1
|
Zhang Z, Zou Y, Song C, Cao K, Cai K, Chen S, Wu Y, Geng D, Sun G, Zhang N, Zhang X, Zhang Y, Sun Y, Zhang Y. Advances in the study of exosomes in cardiovascular diseases. J Adv Res 2024; 66:133-153. [PMID: 38123019 DOI: 10.1016/j.jare.2023.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Cardiovascular disease (CVD) has been the leading cause of death worldwide for many years. In recent years, exosomes have gained extensive attention in the cardiovascular system due to their excellent biocompatibility. Studies have extensively researched miRNAs in exosomes and found that they play critical roles in various physiological and pathological processes in the cardiovascular system. These processes include promoting or inhibiting inflammatory responses, promoting angiogenesis, participating in cell proliferation and migration, and promoting pathological progression such as fibrosis. AIM OF REVIEW This systematic review examines the role of exosomes in various cardiovascular diseases such as atherosclerosis, myocardial infarction, ischemia-reperfusion injury, heart failure and cardiomyopathy. It also presents the latest treatment and prevention methods utilizing exosomes. The study aims to provide new insights and approaches for preventing and treating cardiovascular diseases by exploring the relationship between exosomes and these conditions. Furthermore, the review emphasizes the potential clinical use of exosomes as biomarkers for diagnosing cardiovascular diseases. KEY SCIENTIFIC CONCEPTS OF REVIEW Exosomes are nanoscale vesicles surrounded by lipid bilayers that are secreted by most cells in the body. They are heterogeneous, varying in size and composition, with a diameter typically ranging from 40 to 160 nm. Exosomes serve as a means of information communication between cells, carrying various biologically active substances, including lipids, proteins, and small RNAs such as miRNAs and lncRNAs. As a result, they participate in both physiological and pathological processes within the body.
Collapse
Affiliation(s)
- Zhaobo Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Yuanming Zou
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Chunyu Song
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Kexin Cao
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Kexin Cai
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Shuxian Chen
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Yanjiao Wu
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Danxi Geng
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Guozhe Sun
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Naijin Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China; Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China; Key Laboratory of Reproductive and Genetic Medicine, China Medical University, National Health Commission, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
| | - Xingang Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Yixiao Zhang
- Department of Urology Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, People's Republic of China.
| | - Yingxian Sun
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China; Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
| | - Ying Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China; Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
| |
Collapse
|
2
|
Wu Y, Chen Z, Zheng Z, Li X, Shu J, Mao R, An J, Fan S, Luo R, Guo Y, Xu W, Liang M, Huang K, Wang C. Tudor-SN exacerbates pathological vascular remodeling by promoting the polyubiquitination of PTEN via NEDD4-1. J Biomed Sci 2024; 31:88. [PMID: 39237902 PMCID: PMC11378411 DOI: 10.1186/s12929-024-01076-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 08/20/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Dysregulation of vascular homeostasis can induce cardiovascular diseases and increase global mortality rates. Although lineage tracing studies have confirmed the pivotal role of modulated vascular smooth muscle cells (VSMCs) in the progression of pathological vascular remodeling, the underlying mechanisms are still unclear. METHODS The expression of Tudor-SN was determined in VSMCs of artery stenosis, PDGF-BB-treated VSMCs and atherosclerotic plaque. Loss- and gain-of-function approaches were used to explore the role of Tudor-SN in the modulation of VSMCs phenotype both in vivo and in vitro. RESULTS In this study, we demonstrate that Tudor-SN expression is significantly elevated in injury-induced arteries, atherosclerotic plaques, and PDGF-BB-stimulated VSMCs. Tudor-SN deficiency attenuates, but overexpression aggravates the synthetic phenotypic switching of VSMCs and pathological vascular remodeling. Loss of Tudor-SN also reduces atherosclerotic plaque formation and increases plaque stability. Mechanistically, PTEN, the major regulator of the MAPK and PI3K-AKT signaling pathways, plays a vital role in Tudor-SN-mediated regulation on proliferation and migration of VSMCs. Tudor-SN facilitates the polyubiquitination and degradation of PTEN via NEDD4-1, thus exacerbating vascular remodeling under pathological conditions. BpV (HOpic), a specific inhibitor of PTEN, not only counteracts the protective effect of Tudor-SN deficiency on proliferation and migration of VSMCs, but also abrogates the negative effect of carotid artery injury-induced vascular remodeling in mice. CONCLUSIONS Our findings reveal that Tudor-SN deficiency significantly ameliorated pathological vascular remodeling by reducing NEDD4-1-dependent PTEN polyubiquitination, suggesting that Tudor-SN may be a novel target for preventing vascular diseases.
Collapse
Affiliation(s)
- Yichen Wu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Wuhan, China
- Hubei Clinical Research Center for Metabolic and Cardiovascular Disease, Wuhan, China
| | - Zilong Chen
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Zhe Zheng
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xiaoguang Li
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Jiangcheng Shu
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Ruiqi Mao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Jie An
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Siyuan Fan
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Ruijie Luo
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Yi Guo
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Wenjing Xu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
| | - Minglu Liang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Wuhan, China
- Hubei Clinical Research Center for Metabolic and Cardiovascular Disease, Wuhan, China
| | - Kai Huang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China.
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Wuhan, China.
- Hubei Clinical Research Center for Metabolic and Cardiovascular Disease, Wuhan, China.
| | - Cheng Wang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
- Department of Rheumatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China.
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Wuhan, China.
- Hubei Clinical Research Center for Metabolic and Cardiovascular Disease, Wuhan, China.
| |
Collapse
|
3
|
Zhou P, Meng X, Nie Z, Wang H, Wang K, Du A, Lei Y. PTEN: an emerging target in rheumatoid arthritis? Cell Commun Signal 2024; 22:246. [PMID: 38671436 PMCID: PMC11046879 DOI: 10.1186/s12964-024-01618-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is a critical tumor suppressor protein that regulates various biological processes such as cell proliferation, apoptosis, and inflammatory responses by controlling the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (PI3K/AKT) signaling pathway. PTEN plays a crucial role in the pathogenesis of rheumatoid arthritis (RA). Loss of PTEN may contribute to survival, proliferation, and pro-inflammatory cytokine release of fibroblast-like synoviocytes (FLS). Also, persistent PI3K signaling increases myeloid cells' osteoclastic potential, enhancing localized bone destruction. Recent studies have shown that the expression of PTEN protein in the synovial lining of RA patients with aggressive FLS is minimal. Experimental upregulation of PTEN protein expression could reduce the damage caused by RA. Nonetheless, a complete comprehension of aberrant PTEN drives RA progression and its interactions with other crucial molecules remains elusive. This review is dedicated to promoting a thorough understanding of the signaling mechanisms of aberrant PTEN in RA and aims to furnish pertinent theoretical support for forthcoming endeavors in both basic and clinical research within this domain.
Collapse
Affiliation(s)
- Pan Zhou
- Chengdu Rheumatology Hospital, Chengdu, Sichuan Province, China
| | - Xingwen Meng
- Chengdu Rheumatology Hospital, Chengdu, Sichuan Province, China
| | - Zhimin Nie
- Chengdu Rheumatology Hospital, Chengdu, Sichuan Province, China
| | - Hua Wang
- Chengdu Rheumatology Hospital, Chengdu, Sichuan Province, China
| | - Kaijun Wang
- Nanjing Tongshifeng Hospital, Nanjing, Jiangsu Province, China
| | - Aihua Du
- Zhengzhou Gout and Rheumatology Hospital, Zhengzhou, Henan Province, China
| | - Yu Lei
- Chengdu Rheumatology Hospital, Chengdu, Sichuan Province, China.
| |
Collapse
|
4
|
Orozco-García E, van Meurs DJ, Calderón JC, Narvaez-Sanchez R, Harmsen MC. Endothelial plasticity across PTEN and Hippo pathways: A complex hormetic rheostat modulated by extracellular vesicles. Transl Oncol 2023; 31:101633. [PMID: 36905871 PMCID: PMC10020115 DOI: 10.1016/j.tranon.2023.101633] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/20/2022] [Accepted: 01/25/2023] [Indexed: 03/11/2023] Open
Abstract
Vascularization is a multifactorial and spatiotemporally regulated process, essential for cell and tissue survival. Vascular alterations have repercussions on the development and progression of diseases such as cancer, cardiovascular diseases, and diabetes, which are the leading causes of death worldwide. Additionally, vascularization continues to be a challenge for tissue engineering and regenerative medicine. Hence, vascularization is the center of interest for physiology, pathophysiology, and therapeutic processes. Within vascularization, phosphatase and tensin homolog deleted on chromosome 10 (PTEN) and Hippo signaling have pivotal roles in the development and homeostasis of the vascular system. Their suppression is related to several pathologies, including developmental defects and cancer. Non-coding RNAs (ncRNAs) are among the regulators of PTEN and/or Hippo pathways during development and disease. The purpose of this paper is to review and discuss the mechanisms by which exosome-derived ncRNAs modulate endothelial cell plasticity during physiological and pathological angiogenesis, through the regulation of PTEN and Hippo pathways, aiming to establish new perspectives on cellular communication during tumoral and regenerative vascularization.
Collapse
Affiliation(s)
- Elizabeth Orozco-García
- Physiology and biochemistry research group - PHYSIS, Faculty of Medicine, University of Antioquia, Colombia; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1 (EA11), Groningen 9713 GZ, The Netherlands
| | - D J van Meurs
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1 (EA11), Groningen 9713 GZ, The Netherlands
| | - J C Calderón
- Physiology and biochemistry research group - PHYSIS, Faculty of Medicine, University of Antioquia, Colombia
| | - Raul Narvaez-Sanchez
- Physiology and biochemistry research group - PHYSIS, Faculty of Medicine, University of Antioquia, Colombia
| | - M C Harmsen
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1 (EA11), Groningen 9713 GZ, The Netherlands.
| |
Collapse
|
5
|
Tamayo SO, Cupitra NI, Narvaez-Sanchez R. Vascular adaptation to cancer beyond angiogenesis: The role of PTEN. Microvasc Res 2023; 147:104492. [PMID: 36709859 DOI: 10.1016/j.mvr.2023.104492] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 07/06/2022] [Accepted: 01/24/2023] [Indexed: 01/28/2023]
Abstract
Cancer is a public health problem, and it needs blood vessels to grow. Knowing more about the processes of vascular adaptation to cancer improves our chances of attacking it, since the tumor for its extension needs such adaptation to satisfy its progressive demand for nutrients. The main objective of this review is to present the reader with some fundamental molecular pathways for vascular adaptation to cancer, highlighting within them the regulatory role of homologous tensin and phosphatase protein (PTEN). Hence the review describes vascular adaptation to cancer through somewhat known processes, such as angiogenesis, but emphasizes others that are much less explored, namely the changes in vascular reactivity and remodeling of the vascular wall -intima-media thickness and adjustments in the extracellular matrix- The role of PTEN in physiological and pathological vascular mechanisms in different types of cancer is deepened, as a crucial mediator in vascular adaptation to cancer, and points pending further exploration in cancer vascularization are suggested.
Collapse
Affiliation(s)
- Sofia Ortiz Tamayo
- Physiology and Biochemistry Research Group, PHYSIS, Faculty of Medicine, University of Antioquia, Medellin, Colombia
| | - Nelson Ivan Cupitra
- Physiology and Biochemistry Research Group, PHYSIS, Faculty of Medicine, University of Antioquia, Medellin, Colombia
| | - Raul Narvaez-Sanchez
- Physiology and Biochemistry Research Group, PHYSIS, Faculty of Medicine, University of Antioquia, Medellin, Colombia.
| |
Collapse
|
6
|
Liu S, Liu Y, Liu Z, Hu Y, Jiang M. A review of the signaling pathways of aerobic and anaerobic exercise on atherosclerosis. J Cell Physiol 2023; 238:866-879. [PMID: 36890781 DOI: 10.1002/jcp.30989] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/18/2023] [Accepted: 02/22/2023] [Indexed: 03/10/2023]
Abstract
Atherosclerosis (AS), a chronic inflammatory vascular disease with lipid metabolism abnormalities, is one of the major pathological bases of coronary heart disease. As people's lifestyles and diets change, the incidence of AS increases yearly. Physical activity and exercise training have recently been identified as effective strategies for lowering cardiovascular disease (CVD) risk. However, the best exercise mode to ameliorate the risk factors related to AS is not clear. The effect of exercise on AS is affected by the type of exercise, intensity, and duration. In particular, aerobic and anaerobic exercise are the two most widely discussed types of exercise. During exercise, the cardiovascular system undergoes physiological changes via various signaling pathways. The review aims to summarize signaling pathways related to AS in two different exercise types and provide new ideas for the prevention and treatment of AS in clinical practice.
Collapse
Affiliation(s)
- Sibo Liu
- The QUEEN MARY School, Nanchang University, Nanchang, China
| | - Yuhe Liu
- Medical Collage of Hebei University of Engineering, Handan, China
| | - Zhihan Liu
- The QUEEN MARY School, Nanchang University, Nanchang, China
| | - Yansong Hu
- The QUEEN MARY School, Nanchang University, Nanchang, China
| | - Meixiu Jiang
- The Institute of Translational Medicine, Nanchang University, Nanchang, China
| |
Collapse
|
7
|
Jiang Y, Qian HY. Transcription factors: key regulatory targets of vascular smooth muscle cell in atherosclerosis. Mol Med 2023; 29:2. [PMID: 36604627 PMCID: PMC9817296 DOI: 10.1186/s10020-022-00586-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 12/05/2022] [Indexed: 01/07/2023] Open
Abstract
Atherosclerosis (AS), leading to gradual occlusion of the arterial lumen, refers to the accumulation of lipids and inflammatory debris in the arterial wall. Despite therapeutic advances over past decades including intervention or surgery, atherosclerosis is still the most common cause of cardiovascular diseases and the main mechanism of death and disability worldwide. Vascular smooth muscle cells (VSMCs) play an imperative role in the occurrence of atherosclerosis and throughout the whole stages. In the past, there was a lack of comprehensive understanding of VSMCs, but the development of identification technology, including in vivo single-cell sequencing technology and lineage tracing with the CreERT2-loxP system, suggests that VSMCs have remarkable plasticity and reevaluates well-established concepts about the contribution of VSMCs. Transcription factors, a kind of protein molecule that specifically recognizes and binds DNA upstream promoter regions or distal enhancer DNA elements, play a key role in the transcription initiation of the coding genes and are necessary for RNA polymerase to bind gene promoters. In this review, we highlight that, except for environmental factors, VSMC genes are transcriptionally regulated through complex interactions of multiple conserved cis-regulatory elements and transcription factors. In addition, through a series of transcription-related regulatory processes, VSMCs could undergo phenotypic transformation, proliferation, migration, calcification and apoptosis. Finally, enhancing or inhibiting transcription factors can regulate the development of atherosclerotic lesions, and the downstream molecular mechanism of transcriptional regulation has also been widely studied.
Collapse
Affiliation(s)
- Yu Jiang
- grid.506261.60000 0001 0706 7839Center for Coronary Heart Disease, Department of Cardiology, Fu Wai Hospital, National Center for Cardiovascular Diseases of China, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Rd, Beijing, 100037 China
| | - Hai-Yan Qian
- grid.506261.60000 0001 0706 7839Center for Coronary Heart Disease, Department of Cardiology, Fu Wai Hospital, National Center for Cardiovascular Diseases of China, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Rd, Beijing, 100037 China
| |
Collapse
|
8
|
Ravi Y, Sai-Sudhakar CB, Kuppusamy P. PTEN as a Therapeutic Target in Pulmonary Hypertension Secondary to Left-heart Failure: Effect of HO-3867 and Supplemental Oxygenation. Cell Biochem Biophys 2021; 79:593-607. [PMID: 34133009 DOI: 10.1007/s12013-021-01010-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2021] [Indexed: 01/27/2023]
Abstract
Pulmonary hypertension (PH) is a condition when the pressure in the lung blood vessels is elevated. This leads to increase in thickness of the blood vessels and increases the workload of the heart and lungs. The incidence and prevalence of PH has been on the increase in the last decade. It is estimated that PH affects about 1% of the global population and about 10% of individuals >65 years of age. Of the various types, Group 2 PH is the most common type seen in the elderly population. Fixed PH or PH refractive to therapies is considered a contraindication for heart transplantation; the 30-day mortality in heart transplant recipients is significantly increased in the subset of this population. In general, the pathobiology of PH involves multiple factors including hypoxia, oxidative stress, growth factor receptors, vascular stress, etc. Hence, it is challenging and important to identify specific mechanisms, diagnosis and develop effective therapeutic strategies. The focus of this manuscript is to review some of the important pathobiological processes and mechanisms in the development of PH. Results from our previously reported studies, including targeted treatments along with some new data on PH secondary to left-heart failure, are presented.
Collapse
Affiliation(s)
- Yazhini Ravi
- Department of Surgery, University of Tennessee Health Science Center, Memphis, TN, USA
| | | | - Periannan Kuppusamy
- Departments of Radiology and Medicine, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA.
| |
Collapse
|
9
|
Wang WX, Springer JE, Hatton KW. MicroRNAs as Biomarkers for Predicting Complications following Aneurysmal Subarachnoid Hemorrhage. Int J Mol Sci 2021; 22:ijms22179492. [PMID: 34502401 PMCID: PMC8431281 DOI: 10.3390/ijms22179492] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/27/2021] [Accepted: 08/29/2021] [Indexed: 12/14/2022] Open
Abstract
Aneurysmal subarachnoid hemorrhage (aSAH) is a high mortality hemorrhagic stroke that affects nearly 30,000 patients annually in the United States. Approximately 30% of aSAH patients die during initial hospitalization and those who survive often carry poor prognosis with one in five having permanent physical and/or cognitive disabilities. The poor outcome of aSAH can be the result of the initial catastrophic event or due to the many acute or delayed neurological complications, such as cerebral ischemia, hydrocephalus, and re-bleeding. Unfortunately, no effective biomarker exists to predict or diagnose these complications at a clinically relevant time point when neurologic injury can be effectively treated and managed. Recently, a number of studies have demonstrated that microRNAs (miRNAs) in extracellular biofluids are highly associated with aSAH and complications. Here we provide an overview of the current research on relevant human studies examining the correlation between miRNAs and aSAH complications and discuss the potential application of using miRNAs as biomarkers in aSAH management.
Collapse
Affiliation(s)
- Wang-Xia Wang
- Sanders-Brown Center on Aging, Spinal Cord and Brain Injury Research Center, and the Pathology & Laboratory Medicine, University of Kentucky, Lexington, KY 40536, USA
- Correspondence: ; Tel.: +1-859-218-3886
| | - Joe E. Springer
- Spinal Cord and Brain Injury Research Center, and the Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA;
| | - Kevin W. Hatton
- Department of Anesthesiology Critical Care Medicine, University of Kentucky, Lexington, KY 40536, USA;
| |
Collapse
|
10
|
Maheronnaghsh M, Niktab I, Enayati S, Amoli MM, Hosseini SK, Tavakkoly-Bazzaz J. Differentially expressed miR-152, a potential biomarker for in-stent restenosis (ISR) in peripheral blood mononuclear cells (PBMCs) of coronary artery disease (CAD) patients. Nutr Metab Cardiovasc Dis 2021; 31:1137-1147. [PMID: 33712363 DOI: 10.1016/j.numecd.2020.09.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 09/24/2020] [Accepted: 09/25/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS In-stent restenosis (ISR) remains the most daunting challenge of current treatments of coronary artery disease (CAD). MicroRNAs (miRNAs) are prominent regulators of key pathological processes leading to restenosis and used as diagnostic tools in different studies. miR-152 and miR-148a are implicated to contribute in the putative intracellular mechanisms of ISR. The aim of present study is to investigate the potential early-stage diagnostic role of miR-152 and miR-148a expression levels for ISR in peripheral blood mononuclear cells (PBMCs) of patients who underwent stent implantation. METHODS AND RESULTS The miRNAs that are supposed to be involved in the ISR were nominated by bioinformatics approach mainly using miRWalk3. Then by quantitative real-time PCR, we determined the relative expression of miR-152 and miR-148a of PBMCs from ISR patients with their age/sex-matched controls. RESULTS The presence of ISR significantly coincided with a decrease in the relative expression of miR-152. The area under the curve (AUC) for miR-152 receiver operating characteristic (ROC) curve was 0.717 (95% CI; 0.60-0.83) with a sensitivity of 70% and a specificity of 67%, suggesting that the miRNA expression level might be employed to identify patients at risk of ISR. CONCLUSIONS To the best of our knowledge, this is the first work to show that the miR-152 expression level can possibly be applied to predict CAD patients at risk of ISR. The results suggest that the expression levels of miR-152 in PBMCs may serve as a biomarker for ISR. Our finding suggests the importance of miRNA levels in PBMCs as a novel biological tool to detect diseases in their early clinical stages.
Collapse
Affiliation(s)
- M Maheronnaghsh
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - I Niktab
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - S Enayati
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - M M Amoli
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - S K Hosseini
- Department of Cardiovascular Disorders, Division of Interventional Cardiology, Tehran University of Medical Sciences, Tehran, Iran.
| | - J Tavakkoly-Bazzaz
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Preclinical Investigation of Trifluoperazine as a Novel Therapeutic Agent for the Treatment of Pulmonary Arterial Hypertension. Int J Mol Sci 2021; 22:ijms22062919. [PMID: 33805714 PMCID: PMC7998101 DOI: 10.3390/ijms22062919] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/17/2022] Open
Abstract
Trifluoperazine (TFP), an antipsychotic drug approved by the Food and Drug Administration, has been show to exhibit anti-cancer effects. Pulmonary arterial hypertension (PAH) is a devastating disease characterized by a progressive obliteration of small pulmonary arteries (PAs) due to exaggerated proliferation and resistance to apoptosis of PA smooth muscle cells (PASMCs). However, the therapeutic potential of TFP for correcting the cancer-like phenotype of PAH-PASMCs and improving PAH in animal models remains unknown. PASMCs isolated from PAH patients were exposed to different concentrations of TFP before assessments of cell proliferation and apoptosis. The in vivo therapeutic potential of TFP was tested in two preclinical models with established PAH, namely the monocrotaline and sugen/hypoxia-induced rat models. Assessments of hemodynamics by right heart catheterization and histopathology were conducted. TFP showed strong anti-survival and anti-proliferative effects on cultured PAH-PASMCs. Exposure to TFP was associated with downregulation of AKT activity and nuclear translocation of forkhead box protein O3 (FOXO3). In both preclinical models, TFP significantly lowered the right ventricular systolic pressure and total pulmonary resistance and improved cardiac function. Consistently, TFP reduced the medial wall thickness of distal PAs. Overall, our data indicate that TFP could have beneficial effects in PAH and support the view that seeking new uses for old drugs may represent a fruitful approach.
Collapse
|
12
|
Lin R, Lv J, Wang L, Li X, Zhang J, Sun W, Hu X, Xin S. Potential Target miR-455 Delaying Arterial Stenosis Progression Through PTEN. Front Cardiovasc Med 2021; 8:611116. [PMID: 33708803 PMCID: PMC7940831 DOI: 10.3389/fcvm.2021.611116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 01/06/2021] [Indexed: 11/25/2022] Open
Abstract
Background: Vascular smooth muscle cells (VSMC) underwent phenotypic switching upon stimulation signals, and this is the prerequisite for their proliferation and migration. Previous work revealed that miR-455 may be involved in vascular stenosis. Thus, this study aimed to explore potential targets and mechanisms underlying the dynamics of miR-455 in vascular stenosis. Methods: miR-455 and PTEN expression levels were studied in normal and stenosis tissue, as well as in VSMC in proliferation model. Manipulating miR-455 expression levels was achieved by transfection of either miR-455 mimic or inhibitor, and its effect on cell proliferation was studied by CCK-8 assay. Its effect on gene expression was studied by RT-qPCR and western blot. The expression regulation mechanism was studied by luciferase reporter system. Finally, the effect of miR-455 on regulating vascular stenosis was studied using a rat balloon-injured carotid artery stenosis model. Results: High expression levels of miR-455 were detected in both stenosis arterial tissues and VSMC proliferation models. In contrast, the expression levels of PTEN were downregulated in these systems. miR-455 transfected VSMC showed higher levels of proliferation and decreased levels of PTEN. Potential binding sites between miR-455 and PTEN 3′UTR were predicted and confirmed. NF-kB p65 was found to bind directly on miR-455 promoter region and regulate its transcription. The progression of arterial stenosis could be delayed by introducing miR-455 antagomir. Conclusions: The p65/miR-455/PTEN signaling pathway plays a crucial role in regulating VSMC proliferation and vascular stenosis. This indicated that miR-455 is a novel target that would help improve treatment outcomes in patients suffering from vascular stenosis.
Collapse
Affiliation(s)
- Ruoran Lin
- Department of Vascular Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Junyuan Lv
- Department of Breast and Thyroid Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Lei Wang
- Department of Vascular Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Xuan Li
- Department of Vascular Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Jing Zhang
- Liaoning Key Laboratory of Molecular Tumor Drug Development and Evaluation, Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Weifeng Sun
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaoyun Hu
- Liaoning Key Laboratory of Molecular Tumor Drug Development and Evaluation, Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Shijie Xin
- Department of Vascular Surgery, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
13
|
mTOR Signaling in Pulmonary Vascular Disease: Pathogenic Role and Therapeutic Target. Int J Mol Sci 2021; 22:ijms22042144. [PMID: 33670032 PMCID: PMC7926633 DOI: 10.3390/ijms22042144] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/13/2021] [Accepted: 02/15/2021] [Indexed: 12/16/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive and fatal disease without a cure. The exact pathogenic mechanisms of PAH are complex and poorly understood, yet a number of abnormally expressed genes and regulatory pathways contribute to sustained vasoconstriction and vascular remodeling of the distal pulmonary arteries. Mammalian target of rapamycin (mTOR) is one of the major signaling pathways implicated in regulating cell proliferation, migration, differentiation, and protein synthesis. Here we will describe the canonical mTOR pathway, structural and functional differences between mTOR complexes 1 and 2, as well as the crosstalk with other important signaling cascades in the development of PAH. The pathogenic role of mTOR in pulmonary vascular remodeling and sustained vasoconstriction due to its contribution to proliferation, migration, phenotypic transition, and gene regulation in pulmonary artery smooth muscle and endothelial cells will be discussed. Despite the progress in our elucidation of the etiology and pathogenesis of PAH over the two last decades, there is a lack of effective therapeutic agents to treat PAH patients representing a significant unmet clinical need. In this review, we will explore the possibility and therapeutic potential to use inhibitors of mTOR signaling cascade to treat PAH.
Collapse
|
14
|
Lu S, Jolly AJ, Strand KA, Dubner AM, Mutryn MF, Moulton KS, Nemenoff RA, Majesky MW, Weiser-Evans MC. Smooth muscle-derived progenitor cell myofibroblast differentiation through KLF4 downregulation promotes arterial remodeling and fibrosis. JCI Insight 2020; 5:139445. [PMID: 33119549 PMCID: PMC7714399 DOI: 10.1172/jci.insight.139445] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 10/22/2020] [Indexed: 12/12/2022] Open
Abstract
Resident vascular adventitial SCA1+ progenitor (AdvSca1) cells are essential in vascular development and injury. However, the heterogeneity of AdvSca1 cells presents a unique challenge in understanding signaling pathways orchestrating their behavior in homeostasis and injury responses. Using smooth muscle cell (SMC) lineage-tracing models, we identified a subpopulation of AdvSca1 cells (AdvSca1-SM) originating from mature SMCs that undergo reprogramming in situ and exhibit a multipotent phenotype. Here we employed lineage tracing and RNA-sequencing to define the signaling pathways regulating SMC-to-AdvSca1-SM cell reprogramming and AdvSca1-SM progenitor cell phenotype. Unbiased hierarchical clustering revealed that genes related to hedgehog/WNT/beta-catenin signaling were significantly enriched in AdvSca1-SM cells, emphasizing the importance of this signaling axis in the reprogramming event. Leveraging AdvSca1-SM–specific expression of GLI-Kruppel family member GLI1 (Gli1), we generated Gli1-CreERT2-ROSA26-YFP reporter mice to selectively track AdvSca1-SM cells. We demonstrated that physiologically relevant vascular injury or AdvSca1-SM cell–specific Kruppel-like factor 4 (Klf4) depletion facilitated the proliferation and differentiation of AdvSca1-SM cells to a profibrotic myofibroblast phenotype rather than macrophages. Surprisingly, AdvSca1-SM cells selectively contributed to adventitial remodeling and fibrosis but little to neointima formation. Together, these findings strongly support therapeutics aimed at preserving the AdvSca1-SM cell phenotype as a viable antifibrotic approach. Smooth muscle cell–derived resident vascular adventitial progenitor cells adopt a myofibroblast phenotype in response to vascular injury and play a dominant role in vascular fibrosis.
Collapse
Affiliation(s)
- Sizhao Lu
- Division of Renal Diseases and Hypertension, Department of Medicine, and
| | - Austin J Jolly
- Division of Renal Diseases and Hypertension, Department of Medicine, and
| | - Keith A Strand
- Division of Renal Diseases and Hypertension, Department of Medicine, and
| | - Allison M Dubner
- Division of Renal Diseases and Hypertension, Department of Medicine, and
| | - Marie F Mutryn
- Division of Renal Diseases and Hypertension, Department of Medicine, and
| | | | - Raphael A Nemenoff
- Division of Renal Diseases and Hypertension, Department of Medicine, and.,Consortium for Fibrosis Research and Translation, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Mark W Majesky
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington, USA.,Department of Pediatrics and Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Mary Cm Weiser-Evans
- Division of Renal Diseases and Hypertension, Department of Medicine, and.,Consortium for Fibrosis Research and Translation, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Cardio Vascular Pulmonary Research Lab, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
15
|
Bruijn LE, van den Akker BEWM, van Rhijn CM, Hamming JF, Lindeman JHN. Extreme Diversity of the Human Vascular Mesenchymal Cell Landscape. J Am Heart Assoc 2020; 9:e017094. [PMID: 33190596 PMCID: PMC7763765 DOI: 10.1161/jaha.120.017094] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 10/05/2020] [Indexed: 12/17/2022]
Abstract
Background Human mesenchymal cells are culprit factors in vascular (patho)physiology and are hallmarked by phenotypic and functional heterogeneity. At present, they are subdivided by classic umbrella terms, such as "fibroblasts," "myofibroblasts," "smooth muscle cells," "fibrocytes," "mesangial cells," and "pericytes." However, a discriminative marker-based subclassification has to date not been established. Methods and Results As a first effort toward a classification scheme, a systematic literature search was performed to identify the most commonly used phenotypical and functional protein markers for characterizing and classifying vascular mesenchymal cell subpopulation(s). We next applied immunohistochemistry and immunofluorescence to inventory the expression pattern of identified markers on human aorta specimens representing early, intermediate, and end stages of human atherosclerotic disease. Included markers comprise markers for mesenchymal lineage (vimentin, FSP-1 [fibroblast-specific protein-1]/S100A4, cluster of differentiation (CD) 90/thymocyte differentiation antigen 1, and FAP [fibroblast activation protein]), contractile/non-contractile phenotype (α-smooth muscle actin, smooth muscle myosin heavy chain, and nonmuscle myosin heavy chain), and auxiliary contractile markers (h1-Calponin, h-Caldesmon, Desmin, SM22α [smooth muscle protein 22α], non-muscle myosin heavy chain, smooth muscle myosin heavy chain, Smoothelin-B, α-Tropomyosin, and Telokin) or adhesion proteins (Paxillin and Vinculin). Vimentin classified as the most inclusive lineage marker. Subset markers did not separate along classic lines of smooth muscle cell, myofibroblast, or fibroblast, but showed clear temporal and spatial diversity. Strong indications were found for presence of stem cells/Endothelial-to-Mesenchymal cell Transition and fibrocytes in specific aspects of the human atherosclerotic process. Conclusions This systematic evaluation shows a highly diverse and dynamic landscape for the human vascular mesenchymal cell population that is not captured by the classic nomenclature. Our observations stress the need for a consensus multiparameter subclass designation along the lines of the cluster of differentiation classification for leucocytes.
Collapse
Affiliation(s)
- Laura E. Bruijn
- Division of Vascular SurgeryDepartment of SurgeryLeiden University Medical CenterLeidenthe Netherlands
| | | | - Connie M. van Rhijn
- Division of Vascular SurgeryDepartment of SurgeryLeiden University Medical CenterLeidenthe Netherlands
| | - Jaap F. Hamming
- Division of Vascular SurgeryDepartment of SurgeryLeiden University Medical CenterLeidenthe Netherlands
| | - Jan H. N. Lindeman
- Division of Vascular SurgeryDepartment of SurgeryLeiden University Medical CenterLeidenthe Netherlands
| |
Collapse
|
16
|
Lu YB, Shi C, Yang B, Lu ZF, Wu YL, Zhang RY, He X, Li LM, Hu B, Hu YW, Zheng L, Wang Q. Long noncoding RNA ZNF800 suppresses proliferation and migration of vascular smooth muscle cells by upregulating PTEN and inhibiting AKT/mTOR/HIF-1α signaling. Atherosclerosis 2020; 312:43-53. [PMID: 32971395 DOI: 10.1016/j.atherosclerosis.2020.09.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 06/19/2020] [Accepted: 09/08/2020] [Indexed: 01/31/2023]
Abstract
BACKGROUND AND AIMS Long noncoding RNAs (lncRNAs) have recently been implicated in many biological and disease processes, but the exact mechanism of their involvement in atherosclerosis is unclear. The aberrant proliferation and migration of vascular smooth muscle cells (VSMCs) is a major contributor to the development of atherosclerotic lesions. This study aimed to investigate the potential effects of lncRNA ZNF800, a previously uncharacterized lncRNA, on VSMC proliferation and migration. METHODS The expression of lncRNA ZNF800 in atherosclerotic plaque tissues was detected using reverse transcription-quantitative PCR (RT-qPCR), while the role and mechanism of lncRNA ZNF800 in proliferation and migration of VSMCs were investigated by CCK8 assay, transwell assay, scratch wound assay, RT-qPCR and Western blot. RESULTS We found that lncRNA ZNF800 was significantly more abundant in atherosclerotic plaque tissues, and substantially suppressed the proliferation and migration of VSMCs. LncRNA ZNF800 had no effect on phosphatase and tensin homolog deleted on chromosome 10 (PTEN) mRNA expression but dramatically increased the levels of PTEN protein. Enhanced lncRNA ZNF800 expression inhibited the activity of the AKT/mTOR/HIF-1α signaling pathway, downregulated the expression of vascular endothelial growth factor α (VEGF-α) and matrix metalloproteinase 1 (MMP1), and suppressed VSMC proliferation and migration. These inhibitory effects of lncRNA ZNF800 were abolished by knockdown of PTEN. The inhibitory effects of lncRNA ZNF800 on cell proliferation and migration and the expression of VEGF-α and MMP1 were exacerbated by HIF-1α knockdown in VSMCs. CONCLUSIONS These findings demonstrated that lncRNA ZNF800 suppressed VSMC proliferation and migration by interacting with PTEN through a mechanism involving AKT/mTOR/HIF-1α signaling. Therefore, it may play a key atheroprotective role and represent a potential therapeutic target for atherosclerosis-related diseases.
Collapse
Affiliation(s)
- Yuan-Bin Lu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chao Shi
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Biao Yang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhi-Feng Lu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yi-Lin Wu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ru-Yi Zhang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xin He
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Li-Min Li
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Bing Hu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yan-Wei Hu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Clinical Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, China.
| | - Lei Zheng
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Qian Wang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
17
|
Strand KA, Lu S, Mutryn MF, Li L, Zhou Q, Enyart BT, Jolly AJ, Dubner AM, Moulton KS, Nemenoff RA, Koch KA, LaBarbera DV, Weiser-Evans MCM. High Throughput Screen Identifies the DNMT1 (DNA Methyltransferase-1) Inhibitor, 5-Azacytidine, as a Potent Inducer of PTEN (Phosphatase and Tensin Homolog): Central Role for PTEN in 5-Azacytidine Protection Against Pathological Vascular Remodeling. Arterioscler Thromb Vasc Biol 2020; 40:1854-1869. [PMID: 32580634 DOI: 10.1161/atvbaha.120.314458] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Our recent work demonstrates that PTEN (phosphatase and tensin homolog) is an important regulator of smooth muscle cell (SMC) phenotype. SMC-specific PTEN deletion promotes spontaneous vascular remodeling and PTEN loss correlates with increased atherosclerotic lesion severity in human coronary arteries. In mice, PTEN overexpression reduces plaque area and preserves SMC contractile protein expression in atherosclerosis and blunts Ang II (angiotensin II)-induced pathological vascular remodeling, suggesting that pharmacological PTEN upregulation could be a novel therapeutic approach to treat vascular disease. Approach and Results: To identify novel PTEN activators, we conducted a high-throughput screen using a fluorescence based PTEN promoter-reporter assay. After screening ≈3400 compounds, 11 hit compounds were chosen based on level of activity and mechanism of action. Following in vitro confirmation, we focused on 5-azacytidine, a DNMT1 (DNA methyltransferase-1) inhibitor, for further analysis. In addition to PTEN upregulation, 5-azacytidine treatment increased expression of genes associated with a differentiated SMC phenotype. 5-Azacytidine treatment also maintained contractile gene expression and reduced inflammatory cytokine expression after PDGF (platelet-derived growth factor) stimulation, suggesting 5-azacytidine blocks PDGF-induced SMC de-differentiation. However, these protective effects were lost in PTEN-deficient SMCs. These findings were confirmed in vivo using carotid ligation in SMC-specific PTEN knockout mice treated with 5-azacytidine. In wild type controls, 5-azacytidine reduced neointimal formation and inflammation while maintaining contractile protein expression. In contrast, 5-azacytidine was ineffective in PTEN knockout mice, indicating that the protective effects of 5-azacytidine are mediated through SMC PTEN upregulation. CONCLUSIONS Our data indicates 5-azacytidine upregulates PTEN expression in SMCs, promoting maintenance of SMC differentiation and reducing pathological vascular remodeling in a PTEN-dependent manner.
Collapse
Affiliation(s)
- Keith A Strand
- From the Division of Renal Diseases and Hypertension, Department of Medicine (K.A.S., S.L., M.F.M., A.J.J., A.M.D., R.A.N., M.C.M.W.-E.), University of Colorado, Anschutz Medical Campus, Aurora
| | - Sizhao Lu
- From the Division of Renal Diseases and Hypertension, Department of Medicine (K.A.S., S.L., M.F.M., A.J.J., A.M.D., R.A.N., M.C.M.W.-E.), University of Colorado, Anschutz Medical Campus, Aurora
| | - Marie F Mutryn
- From the Division of Renal Diseases and Hypertension, Department of Medicine (K.A.S., S.L., M.F.M., A.J.J., A.M.D., R.A.N., M.C.M.W.-E.), University of Colorado, Anschutz Medical Campus, Aurora
| | - Linfeng Li
- School of Pharmacy and Pharmaceutical Sciences (L.L., Q.Z., D.V.L.), University of Colorado, Anschutz Medical Campus, Aurora
| | - Qiong Zhou
- School of Pharmacy and Pharmaceutical Sciences (L.L., Q.Z., D.V.L.), University of Colorado, Anschutz Medical Campus, Aurora
| | - Blake T Enyart
- School of Medicine, Consortium for Fibrosis Research & Translation (B.T.E., K.S.M., R.A.N., K.A.K., M.C.M.W.-E.), University of Colorado, Anschutz Medical Campus, Aurora.,Division of Cardiology, Department of Medicine (B.T.E., K.S.M., K.A.K.), University of Colorado, Anschutz Medical Campus, Aurora
| | - Austin J Jolly
- From the Division of Renal Diseases and Hypertension, Department of Medicine (K.A.S., S.L., M.F.M., A.J.J., A.M.D., R.A.N., M.C.M.W.-E.), University of Colorado, Anschutz Medical Campus, Aurora
| | - Allison M Dubner
- From the Division of Renal Diseases and Hypertension, Department of Medicine (K.A.S., S.L., M.F.M., A.J.J., A.M.D., R.A.N., M.C.M.W.-E.), University of Colorado, Anschutz Medical Campus, Aurora
| | - Karen S Moulton
- School of Medicine, Consortium for Fibrosis Research & Translation (B.T.E., K.S.M., R.A.N., K.A.K., M.C.M.W.-E.), University of Colorado, Anschutz Medical Campus, Aurora.,Division of Cardiology, Department of Medicine (B.T.E., K.S.M., K.A.K.), University of Colorado, Anschutz Medical Campus, Aurora
| | - Raphael A Nemenoff
- From the Division of Renal Diseases and Hypertension, Department of Medicine (K.A.S., S.L., M.F.M., A.J.J., A.M.D., R.A.N., M.C.M.W.-E.), University of Colorado, Anschutz Medical Campus, Aurora.,School of Medicine, Consortium for Fibrosis Research & Translation (B.T.E., K.S.M., R.A.N., K.A.K., M.C.M.W.-E.), University of Colorado, Anschutz Medical Campus, Aurora
| | - Keith A Koch
- School of Medicine, Consortium for Fibrosis Research & Translation (B.T.E., K.S.M., R.A.N., K.A.K., M.C.M.W.-E.), University of Colorado, Anschutz Medical Campus, Aurora.,Division of Cardiology, Department of Medicine (B.T.E., K.S.M., K.A.K.), University of Colorado, Anschutz Medical Campus, Aurora
| | - Daniel V LaBarbera
- School of Pharmacy and Pharmaceutical Sciences (L.L., Q.Z., D.V.L.), University of Colorado, Anschutz Medical Campus, Aurora
| | - Mary C M Weiser-Evans
- From the Division of Renal Diseases and Hypertension, Department of Medicine (K.A.S., S.L., M.F.M., A.J.J., A.M.D., R.A.N., M.C.M.W.-E.), University of Colorado, Anschutz Medical Campus, Aurora.,School of Medicine, Consortium for Fibrosis Research & Translation (B.T.E., K.S.M., R.A.N., K.A.K., M.C.M.W.-E.), University of Colorado, Anschutz Medical Campus, Aurora
| |
Collapse
|
18
|
Wu W, Jing D, Meng Z, Hu B, Zhong B, Deng X, Jin X, Shao Z. FGD1 promotes tumor progression and regulates tumor immune response in osteosarcoma via inhibiting PTEN activity. Am J Cancer Res 2020; 10:2859-2871. [PMID: 32194840 PMCID: PMC7052884 DOI: 10.7150/thno.41279] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/11/2020] [Indexed: 12/26/2022] Open
Abstract
Rationale: Mesenchymal cell-derived osteosarcoma is a rare malignant bone tumor affecting children and adolescents. PTEN down-regulation or function-loss mutation is associated with the aggressive of osteosarcoma. Explicating the regulatory mechanism of PTEN might highlight new targets for improving the survival rate of osteosarcoma patients. Methods: The clinical relevance of FGD1 was examined by the TCGA data set, Western blotting and immunohistochemistry of osteosarcoma microarray slides. Functional assays, such as the MTS assay, colony formation assay and xenografts, were used to determine the biological role of FGD1 in osteosarcoma. The protein-protein interaction between FGD1 and PTEN was detected via co-immunoprecipitation. The relationship between FGD1 and PD-L1 was examined by Western blot analysis, RT-qPCR and immunohistochemistry. Results: In this study, analysis of the TCGA data set of sarcomas revealed that FGD1 was over-expressed with the highest P values. Then, we demonstrated that FGD1 was also abnormally up-regulated in osteosarcoma with unfavorable prognosis. Aberrant expressed FGD1 promoted the osteosarcoma tumor cell proliferation and invasion. Moreover, we found that FGD1 was participated in activating PI3K/AKT signaling pathway by interacting with PTEN. Finally, we showed that FGD1 was capable of regulating the tumor immune response via the PTEN/PD-L1 axis in osteosarcoma. Conclusions: Our data suggested that abnormally over-expressed FGD1 functions as an oncogenic protein to promote osteosarcoma progression through inhibiting PTEN activity and activating PI3K/AKT signaling. Notably, FGD1 increased PD-L1 expression in a PTEN dependent manner and modulated the sensitivity of immune checkpoint-based immunotherapy in osteosarcoma. Thus, FGD1 might be a potential target for improving the survival rate of osteosarcomas.
Collapse
|
19
|
Lu S, Strand KA, Mutryn MF, Tucker RM, Jolly AJ, Furgeson SB, Moulton KS, Nemenoff RA, Weiser-Evans MCM. PTEN (Phosphatase and Tensin Homolog) Protects Against Ang II (Angiotensin II)-Induced Pathological Vascular Fibrosis and Remodeling-Brief Report. Arterioscler Thromb Vasc Biol 2019; 40:394-403. [PMID: 31852223 DOI: 10.1161/atvbaha.119.313757] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Pathological vascular remodeling and excessive perivascular fibrosis are major contributors to reduced vessel compliance that exacerbates cardiovascular diseases, for instance, promoting clinically relevant myocardial remodeling. Inflammation plays a significant role in both pathological vascular remodeling and fibrosis. We previously demonstrated that smooth muscle cell-specific PTEN depletion promotes significant vascular fibrosis and accumulation of inflammatory cells. In the current study, we aimed to determine the beneficial role of systemic PTEN elevation on Ang II (angiotensin II)-induced vascular fibrosis and remodeling. Approach and Results: Transgenic mice carrying additional copies of the wild-type Pten gene (super PTEN [sPTEN]) and WT littermates were subjected to Ang II or saline infusion for 14 or 28 days. Compared with WT, Ang II-induced vascular fibrosis was significantly blunted in sPTEN mice, as shown by histochemical stainings and label-free second harmonic generation imaging. The protection against Ang II was recapitulated in sPTEN mice bearing WT bone marrow but not in WT mice reconstituted with sPTEN bone marrow. Ang II-induced elevation of profibrotic and proinflammatory gene expression observed in WT mice was blocked in aortic tissue of sPTEN mice. Immunofluorescent staining and flow cytometry both indicated that perivascular infiltration of T cells and macrophages was significantly inhibited in sPTEN mice. In vitro induction of PTEN expression suppressed Ang II-induced Ccl2 expression in vascular smooth muscle cells. CONCLUSIONS Systemic PTEN elevation mediates protection against Ang II-induced vascular inflammation and fibrosis predominantly through effects in resident vascular cells. Our data highly support that pharmacological upregulation of PTEN could be a novel and viable approach for the treatment of pathological vascular fibrosis.
Collapse
Affiliation(s)
- Sizhao Lu
- From the Division of Renal Diseases and Hypertension, Department of Medicine (S.L., K.A.S., M.F.M., R.M.T., A.J.J., S.B.F., R.A.N., M.C.M.W.-E.), University of Colorado, Anschutz Medical Campus, Aurora
| | - Keith A Strand
- From the Division of Renal Diseases and Hypertension, Department of Medicine (S.L., K.A.S., M.F.M., R.M.T., A.J.J., S.B.F., R.A.N., M.C.M.W.-E.), University of Colorado, Anschutz Medical Campus, Aurora
| | - Marie F Mutryn
- From the Division of Renal Diseases and Hypertension, Department of Medicine (S.L., K.A.S., M.F.M., R.M.T., A.J.J., S.B.F., R.A.N., M.C.M.W.-E.), University of Colorado, Anschutz Medical Campus, Aurora
| | - Rebecca M Tucker
- From the Division of Renal Diseases and Hypertension, Department of Medicine (S.L., K.A.S., M.F.M., R.M.T., A.J.J., S.B.F., R.A.N., M.C.M.W.-E.), University of Colorado, Anschutz Medical Campus, Aurora
| | - Austin J Jolly
- From the Division of Renal Diseases and Hypertension, Department of Medicine (S.L., K.A.S., M.F.M., R.M.T., A.J.J., S.B.F., R.A.N., M.C.M.W.-E.), University of Colorado, Anschutz Medical Campus, Aurora
| | - Seth B Furgeson
- From the Division of Renal Diseases and Hypertension, Department of Medicine (S.L., K.A.S., M.F.M., R.M.T., A.J.J., S.B.F., R.A.N., M.C.M.W.-E.), University of Colorado, Anschutz Medical Campus, Aurora.,School of Medicine, Consortium for Fibrosis Research and Translation (S.B.F., R.A.N., M.C.M.W.-E.), University of Colorado, Anschutz Medical Campus, Aurora
| | - Karen S Moulton
- Division of Cardiology, Department of Medicine (K.S.M.), University of Colorado, Anschutz Medical Campus, Aurora
| | - Raphael A Nemenoff
- From the Division of Renal Diseases and Hypertension, Department of Medicine (S.L., K.A.S., M.F.M., R.M.T., A.J.J., S.B.F., R.A.N., M.C.M.W.-E.), University of Colorado, Anschutz Medical Campus, Aurora.,School of Medicine, Consortium for Fibrosis Research and Translation (S.B.F., R.A.N., M.C.M.W.-E.), University of Colorado, Anschutz Medical Campus, Aurora
| | - Mary C M Weiser-Evans
- From the Division of Renal Diseases and Hypertension, Department of Medicine (S.L., K.A.S., M.F.M., R.M.T., A.J.J., S.B.F., R.A.N., M.C.M.W.-E.), University of Colorado, Anschutz Medical Campus, Aurora.,School of Medicine, Consortium for Fibrosis Research and Translation (S.B.F., R.A.N., M.C.M.W.-E.), University of Colorado, Anschutz Medical Campus, Aurora
| |
Collapse
|
20
|
Wang C, Feng Y, Zhang C, Cheng D, Wu R, Yang Y, Sargsyan D, Kumar D, Kong AN. PTEN deletion drives aberrations of DNA methylome and transcriptome in different stages of prostate cancer. FASEB J 2019; 34:1304-1318. [PMID: 31914691 DOI: 10.1096/fj.201901205rr] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 12/11/2022]
Abstract
Phosphatase and tensin homolog located on chromosome 10 (PTEN) is a tumor suppressor gene and one of the most frequently mutated/deleted genes in human prostate cancer (PCa). However, how PTEN deletion would impact the epigenome and transcriptome alterations remain unknown. This hypothesis was tested in a prostate-specific PTEN-/- (KO) mouse prostatic adenocarcinoma model through DNA methyl-Seq and RNA-Seq analyses. Examination of cancer genomic datasets revealed that PTEN is expressed at lower levels in PTEN-deleted tumor samples than in normal solid tissue samples. Methylome and transcriptome profiling identified several inflammatory responses and immune response signaling pathways, including NF-kB signaling, IL-6 signaling, LPS/IL-1-mediated inhibition of RXR Function, PI3K in B lymphocytes, iCOS-iCOSL in T helper cells, and the role of NFAT in regulating the immune response, were affected by PTEN deletion. Importantly, a small subset of genes that showed DNA hypermethylation or hypomethylation was correlated with decreased or increased gene expression including CXCL1. quantitative polymerase chain reaction analyses of representative genes validated the RNA-Seq results. Histopathological examinations showed that the severity of prostatic intraepithelial neoplasia and inflammation development gradually increased as PTEN null mice aged. Collectively, these findings suggest that loss of PTEN drives global changes in DNA CpG methylation and transcriptomic gene expression and highly associated with several inflammatory and immune molecular pathways during PCa development. These biomarkers could be valuable molecular targets for cancer drug discovery and development against PCa.
Collapse
Affiliation(s)
- Chao Wang
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,Center for Phytochemical Epigenome Studies, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Yaping Feng
- Genomics Core Facility, Waksman Institute of Microbiology, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Chengyue Zhang
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,Center for Phytochemical Epigenome Studies, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - David Cheng
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,Center for Phytochemical Epigenome Studies, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,Graduate Program of Pharmaceutical Sciences, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Renyi Wu
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,Center for Phytochemical Epigenome Studies, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Yuqing Yang
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,Center for Phytochemical Epigenome Studies, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Davit Sargsyan
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,Center for Phytochemical Epigenome Studies, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,Graduate Program of Pharmaceutical Sciences, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Dibyendu Kumar
- Genomics Core Facility, Waksman Institute of Microbiology, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Ah-Ng Kong
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,Center for Phytochemical Epigenome Studies, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| |
Collapse
|
21
|
Zhu J, Liu B, Wang Z, Wang D, Ni H, Zhang L, Wang Y. Exosomes from nicotine-stimulated macrophages accelerate atherosclerosis through miR-21-3p/PTEN-mediated VSMC migration and proliferation. Theranostics 2019; 9:6901-6919. [PMID: 31660076 PMCID: PMC6815950 DOI: 10.7150/thno.37357] [Citation(s) in RCA: 253] [Impact Index Per Article: 50.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 08/06/2019] [Indexed: 12/14/2022] Open
Abstract
Rationale: During the development of atherosclerosis, macrophages secrete exosomes that regulate vascular smooth muscle cells (VSMCs); however, whether nicotine, a major constituent of cigarettes, can modulate this communication in the context of atherogenesis remains to be further studied. In this study, we hypothesized that nicotine induces macrophages to secrete atherogenic exosomes containing microRNAs (miRNAs) to mediate cell-to-cell crosstalk and encourage proatherogenic phenotypes of VSMCs. Methods: In an in vivo study, nicotine was administered subcutaneously to 8-week-old male ApoE-/- mice fed a high-fat diet (HFD) for 12 weeks. Oil red O and hematoxylin and eosin (HE) were used to stain atherosclerotic lesions. Lesion macrophages, VSMCs and exosomes were stained for CD68, α-smooth muscle actin (α-SMA) and CD9, and plaque exosomes were observed by transmission electron microscopy (TEM). Exosomes derived from control macrophages (M-Exos) and from nicotine-treated macrophages (NM-Exos) were isolated by ultracentrifugation, purified by sucrose density gradient centrifugation and characterized based on specific morphology and surface markers. The IVIS® Spectrum in vivo imaging system showed the biodistribution of NM-Exos and M-Exos in circulation. Chitosan hydrogel-incorporated exosomes were applied to simulate exosome secretion in situ. Scratch wound assay, transwell assay and EdU staining were conducted to assess the effects of NM-Exos on the migration and proliferation of mouse VSMCs. RNA-seq was performed to determine the miRNA profiles of M-Exos and NM-Exos. Quantitative real-time PCR (qRT-PCR) analysis was conducted to detect the expression levels of miRNAs and mRNAs. The roles of the candidate miRNA and its target gene were assessed using specific RNA inhibitors, siRNAs and miRNA mimics. Western blotting was used to detect candidate protein expression levels. A dual-luciferase reporting system was utilized to confirm the binding of a specific miRNA to its target gene. Results: Nicotine induced atherosclerotic lesion progression and resulted in plaque exosome retention in vivo. The biodistribution of NM-Exos showed that plaque-resident exosomes might be secreted in situ. VSMCs cocultured in vitro with nicotine-stimulated macrophages presented an increased capacity for migration and proliferation, which was exosome-dependent. In addition, isolated NM-Exos helped promote VSMC migration and proliferation. miRNA profiling showed that miR-21-3p was enriched in NM-Exos, and this miRNA was shown to play a key role in regulating NM-Exos-induced effects by directly targeting phosphatase and tension homologue (PTEN). Conclusion: Exosomal miR-21-3p from nicotine-treated macrophages may accelerate the development of atherosclerosis by increasing VSMC migration and proliferation through its target PTEN.
Collapse
|
22
|
Sundararajan SR, Rajagopalakrishnan R, Rajasekaran S. Arthroscopic Excision of Angio-Fibro-Lipomatous Hamartoma of the Knee: A Rare Case Report. Indian J Orthop 2019; 53:732-735. [PMID: 31673174 PMCID: PMC6804391 DOI: 10.4103/ortho.ijortho_330_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Angio-fibro-lipomatous hamartoma is a benign adipose tissue tumor very rarely seen in musculoskeletal distribution, and its incidence in the knee joint has never been reported. The patient in our case presented with knee pain of 2 years' duration, following blunt trauma. Preoperatively, veno-lymphatic malformation/hemangioma was considered as the diagnosis. Only after arthroscopic excision biopsy, histopathological examination, retrospective radiological analysis, and a review of literature, we were able to diagnose this rare condition. The histopathological picture of this benign adipose tissue tumor contained a mixture of mature adipose tissue and fibrous and vascular tissues. Here, in this case report, we discuss about PTEN gene causing PTEN hamartoma of soft tissue and angiolipoma presentations and its variants.
Collapse
Affiliation(s)
| | - Ramakanth Rajagopalakrishnan
- Department of Arthroscopy and Sports Medicine, Ganga Hospital, Coimbatore, Tamil Nadu, India,Address for correspondence: Dr. Ramakanth Rajagopalakrishnan, Ganga Hospital, Coimbatore, Tamil Nadu, India. E-mail:
| | | |
Collapse
|
23
|
Yu Q, Li W, Xie D, Zheng X, Huang T, Xue P, Guo B, Gao Y, Zhang C, Sun P, Li M, Wang G, Cheng X, Zheng Q, Song Z. PI3Kγ promotes vascular smooth muscle cell phenotypic modulation and transplant arteriosclerosis via a SOX9-dependent mechanism. EBioMedicine 2018; 36:39-53. [PMID: 30241919 PMCID: PMC6197754 DOI: 10.1016/j.ebiom.2018.09.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 08/31/2018] [Accepted: 09/10/2018] [Indexed: 12/11/2022] Open
Abstract
Background Transplant arteriosclerosis (TA) remains the major cause of chronic graft failure in solid organ transplantation. The phenotypic modulation of vascular smooth muscle cells (VSMCs) is a key event for the initiation and progression of neointimal formation and TA. This study aims to explore the role and underlying mechanism of phosphoinositide 3-kinases γ (PI3Kγ) in VSMC phenotypic modulation and TA. Methods The rat model of aortic transplantation was established to detect PI3Kγ expression and its role in neointimal formation and vascular remodeling in vivo. PI3Kγ shRNA transfection was employed to knockdown PI3Kγ gene. Aortic VSMCs was cultured and treated with TNF-α to explore the role and molecular mechanism of PI3Kγ in VSMC phenotypic modulation. Findings Activated PI3Kγ/p-Akt signaling was observed in aortic allografts and in TNF-α-treated VSMCs. Lentivirus-mediated shRNA transfection effectively inhibited PI3Kγ expression in medial VSMCs while restoring the expression of VSMC contractile genes, associated with impaired neointimal formation in aortic allografts. In cultured VSMCs, PI3Kγ blockade with pharmacological inhibitor or genetic knockdown markedly abrogated TNF-α-induced downregulation of VSMC contractile genes and increase in cellular proliferation and migration. Moreover, SOX9 located in nucleus competitively inhibited the interaction of Myocardin and SRF, while PI3Kγ inhibition robustly reduced SOX9 expression and its nuclear translocation and repaired the Myocardin/SRF association. Interpretation These results suggest that PI3Kγ plays a critical role in VSMC phenotypic modulation via a SOX9-dependent mechanism. Therefore, PI3Kγ in VSMCs may represent a promising therapeutic target for the treatment of TA. Fund National Natural Science Foundation of China.
Collapse
Affiliation(s)
- Qihong Yu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Li
- Departments of Gerontology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dawei Xie
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xichuan Zheng
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tong Huang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Xue
- Departments of Gerontology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bing Guo
- Department of Hepatology and Oncology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Yang Gao
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Zhang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Sun
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Li
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guoliang Wang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Cheng
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qichang Zheng
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Zifang Song
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
24
|
Dutzmann J, Koch A, Weisheit S, Sonnenschein K, Korte L, Haertlé M, Thum T, Bauersachs J, Sedding DG, Daniel JM. Sonic hedgehog-dependent activation of adventitial fibroblasts promotes neointima formation. Cardiovasc Res 2018; 113:1653-1663. [PMID: 29088375 DOI: 10.1093/cvr/cvx158] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 08/14/2017] [Indexed: 12/13/2022] Open
Abstract
Aims Adventitial cells have been suggested to contribute to neointima formation, but the functional relevance and the responsible signalling pathways are largely unknown. Sonic hedgehog (Shh) is a regulator of vasculogenesis and promotes angiogenesis in the adult. Methods and results Here we show that proliferation of vascular smooth muscle cells (SMC) after wire-induced injury in C57BL/6 mice is preceded by proliferation of adventitial fibroblasts. Simultaneously, the expression of Shh and its downstream signalling protein smoothened (SMO) were robustly increased within injured arteries. In vitro, combined stimulation with Shh and platelet-derived growth factor (PDGF)-BB strongly induced proliferation and migration of human adventitial fibroblasts. The supernatant of these activated fibroblasts contained high levels of interleukin-6 and -8 and strongly induced proliferation and migration of SMC. Inhibition of SMO selectively prevented fibroblast proliferation, cytokine release, and paracrine SMC activation. Mechanistically, we found that PDGF-BB activates protein kinase A in fibroblasts and thereby induces trafficking of SMO to the plasma membrane, where it can be activated by Shh. In vivo, SMO-inhibition significantly prevented the proliferation of adventitial fibroblasts and neointima formation following wire-induced injury. Conclusions The initial activation of adventitial fibroblasts is essential for the subsequent proliferation of SMC and neointima formation. We identified SMO-dependent Shh signalling as a specific process for the activation of adventitial fibroblasts.
Collapse
Affiliation(s)
- Jochen Dutzmann
- Vascular Remodeling and Regeneration Group, Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Alexander Koch
- Vascular Remodeling and Regeneration Group, Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Simona Weisheit
- Vascular Remodeling and Regeneration Group, Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Kristina Sonnenschein
- Vascular Remodeling and Regeneration Group, Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany.,Institute for Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Laura Korte
- Vascular Remodeling and Regeneration Group, Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Marco Haertlé
- Vascular Remodeling and Regeneration Group, Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Thomas Thum
- Institute for Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany.,National Heart and Lung Institute, Imperial College, Sydney St, Chelsea, London SW3 6NP, UK
| | - Johann Bauersachs
- Vascular Remodeling and Regeneration Group, Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Daniel G Sedding
- Vascular Remodeling and Regeneration Group, Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Jan-Marcus Daniel
- Vascular Remodeling and Regeneration Group, Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| |
Collapse
|
25
|
Yu B, Chen Q, Le Bras A, Zhang L, Xu Q. Vascular Stem/Progenitor Cell Migration and Differentiation in Atherosclerosis. Antioxid Redox Signal 2018; 29:219-235. [PMID: 28537424 DOI: 10.1089/ars.2017.7171] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
SIGNIFICANCE Atherosclerosis is a major cause for the death of human beings, and it takes place in large- and middle-sized arteries. The pathogenesis of the disease has been widely investigated, and new findings on vascular stem/progenitor cells could have an impact on vascular regeneration. Recent Advances: Recent studies have shown that abundant stem/progenitor cells present in the vessel wall are mainly responsible for cell accumulation in the intima during vascular remodeling. It has been demonstrated that the mobilization and recruitment of tissue-resident stem/progenitor cells give rise to endothelial and smooth muscle cells (SMCs) that participate in vascular repair and remodeling such as neointimal hyperplasia and arteriosclerosis. Interestingly, cell lineage tracing studies indicate that a large proportion of SMCs in neointimal lesions is derived from adventitial stem/progenitor cells. CRITICAL ISSUES The influence of stem/progenitor cell behavior on the development of atherosclerosis is crucial. An understanding of the regulatory mechanisms that control stem/progenitor cell migration and differentiation is essential for stem/progenitor cell therapy for vascular diseases and regenerative medicine. FUTURE DIRECTIONS Identification of the detailed process driving the migration and differentiation of vascular stem/progenitor cells during the development of atherosclerosis, discovery of the environmental cues, and signaling pathways that control cell fate within the vasculature will facilitate the development of new preventive and therapeutic strategies to combat atherosclerosis. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Baoqi Yu
- 1 Department of Emergency, Guangdong General Hospital , Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Qishan Chen
- 2 Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, China
| | - Alexandra Le Bras
- 3 Cardiovascular Division, King's College London BHF Centre , London, United Kingdom
| | - Li Zhang
- 2 Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, China
| | - Qingbo Xu
- 3 Cardiovascular Division, King's College London BHF Centre , London, United Kingdom
| |
Collapse
|
26
|
Chen WJ, Chen YH, Hsu YJ, Lin KH, Yeh YH. MicroRNA-132 targeting PTEN contributes to cilostazol-promoted vascular smooth muscle cell differentiation. Atherosclerosis 2018; 274:1-7. [PMID: 29738818 DOI: 10.1016/j.atherosclerosis.2018.04.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 04/09/2018] [Accepted: 04/25/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Cilostazol, beyond its antiplatelet effect, is also capable of promoting vascular smooth muscle cell (VSMC) differentiation. The aim of this study was to explore the potential role of PTEN, known to associate with VSMC differentiation, and its related microRNA (miRNA) in cilostazol-dependent effects. METHODS AND RESULTS Microarray analysis in balloon-injured rat carotid arteries comparing with and without balloon injury revealed that miR-132 was differentially expressed. Bioinformatic analysis predicts PTEN as a novel target of miR-132. Western blot and quantitative real-time reverse transcription-polymerase chain reaction along with in situ hybridization documented that cilostazol treatment enhanced PTEN and reduced miR-132 expression in the neointima of balloon-injured arteries. Treatment of cultured rat VSMCs with cilostazol resulted in the up-regulation of PTEN mRNA and the down-regulation of miR-132, supporting an in vitro relevance. Co-transfection experiments showed that transfection of miR-132 mimic into VSMCs suppressed PTEN 3'UTR activities, further reflecting that PTEN is the direct target of miR-132. Over-expression of miR-132 in VSMCs led to an attenuation of cilostazol-induced PTEN and its downstream VSMC differentiation marker (calponin) expression, confirming the critical role of miR-132 in VSMC differentiation. Transient transfection studies demonstrated that cilostazol reduced the activity of miR-132 promoter, which was mediated via cyclic AMP response element-binding protein. Notably, the use of lentivirus to over-express miR-132 in the neointima of balloon-injured arteries could reverse the effect of cilostazol in vivo. CONCLUSIONS These results suggest that miR-132 by targeting PTEN may be an important regulator in mediating cilostazol actions on VSMC differentiation.
Collapse
MESH Headings
- 3' Untranslated Regions
- Animals
- Binding Sites
- Calcium-Binding Proteins/genetics
- Calcium-Binding Proteins/metabolism
- Cardiovascular Agents/pharmacology
- Carotid Arteries/drug effects
- Carotid Arteries/embryology
- Carotid Arteries/pathology
- Carotid Artery Injuries/drug therapy
- Carotid Artery Injuries/enzymology
- Carotid Artery Injuries/genetics
- Carotid Artery Injuries/pathology
- Cell Differentiation/drug effects
- Cells, Cultured
- Cilostazol/pharmacology
- Cyclic AMP Response Element-Binding Protein/metabolism
- Disease Models, Animal
- Down-Regulation
- Gene Expression Regulation, Enzymologic
- Male
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Microfilament Proteins/genetics
- Microfilament Proteins/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- PTEN Phosphohydrolase/genetics
- PTEN Phosphohydrolase/metabolism
- Promoter Regions, Genetic
- Rats, Wistar
- Signal Transduction/drug effects
- Calponins
Collapse
Affiliation(s)
- Wei-Jan Chen
- Division of Cardiology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Tao-Yuan, Taiwan.
| | - Ying-Hwa Chen
- Division of Cardiology, Department of Internal Medicine, Taipei Veterans General Hospital, National Yang-Ming University College of Medicine, Taipei, Taiwan
| | - Yu-Juei Hsu
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| | - Kwang-Huei Lin
- Department of Biochemistry, College of Medicine, Chang-Gung University, Liver Research Center, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Yung-Hsin Yeh
- Division of Cardiology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Tao-Yuan, Taiwan
| |
Collapse
|
27
|
Wang K, Deng P, Sun Y, Ye P, Zhang A, Wu C, Yue Z, Chen Z, Xia J. MicroRNA-155 promotes neointimal hyperplasia through smooth muscle-like cell-derived RANTES in arteriovenous fistulas. J Vasc Surg 2018; 67:933-944.e3. [PMID: 29477204 DOI: 10.1016/j.jvs.2017.02.046] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 02/19/2017] [Indexed: 10/18/2022]
Abstract
OBJECTIVE Arteriovenous fistula (AVF) suffers from a high number of failures caused by insufficient outward remodeling and venous neointimal hyperplasia formation. The aim was to investigate the exact mechanism by which microRNA-155 (miR-155) in the outflow vein of AVF is regulated. METHODS AVFs between the branch of the jugular vein and carotid artery in an end-to-end manner were created in C57BL/6 and miR-155-/- mice with a C57BL/6 background. The venous segments were harvested at day 7, 14, 21, and 28, and the AVFs were analyzed histologically and at a messenger RNA level using real-time quantitative polymerase chain reactions. The outflow vein of AVF and the normal great saphenous vein, collected from patients with chronic kidney disease and coronary artery bypass surgery, were analyzed by histologic and molecular biologic approaches. RESULTS Venous neointimal hyperplasia is significantly alleviated in miR-155-/- mice, and the expression of several chemokines and cytokines in the vessel wall, including regulated on activation, normal T-cell expressed and secreted factor (RANTES), monocyte chemoattractant protein 1, and vascular endothelial growth factor, was inhibited. miR-155 promoted the RANTES expression of smooth muscle-like cells, which in turn facilitated cell proliferation and extracellular matrix production. CONCLUSIONS miR-155 enhances venous neointima formation through the autocrine and paracrine effects of smooth muscle-like cell-derived RANTES in a nuclear factor κB-dependent manner during the entire AVF process, especially at the advanced stage.
Collapse
Affiliation(s)
- Ke Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Deng
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Sun
- Department of Vascular Surgery, Clinical Medical School of Yangzhou University, Yangzhou, China
| | - Ping Ye
- Department of Cardiovascular Medicine and Department of Cardiovascular Surgery, Central Hospital of Wuhan, Wuhan, China
| | - Anchen Zhang
- Department of Cardiovascular Medicine and Department of Cardiovascular Surgery, Central Hospital of Wuhan, Wuhan, China
| | - Chuangyan Wu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhang Yue
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhaolei Chen
- Department of Vascular Surgery, Clinical Medical School of Yangzhou University, Yangzhou, China
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Cardiovascular Medicine and Department of Cardiovascular Surgery, Central Hospital of Wuhan, Wuhan, China.
| |
Collapse
|
28
|
Guo D, Wang YW, Yan L, Ma J, Han XW, Shui SF. Dysregulation of microRNA‑23b‑3p contributes to the development of intracranial aneurysms by targeting phosphatase and tensin homolog. Int J Mol Med 2018; 42:1637-1643. [PMID: 29845190 DOI: 10.3892/ijmm.2018.3706] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 10/19/2017] [Indexed: 11/06/2022] Open
Abstract
MicroRNA‑23b‑3p (miR‑23b‑3p) has been reported to be involved in the pathogenesis of a number of diseases, including non‑small cell lung cancer and gastric cancer, by acting on different signaling pathways. The present study aimed to understand the association between the miR‑23b‑3p level of intracranial aneurysms (IAs) and the mechanism involved. Computational analysis was used to search for the target of miR‑23b‑3p, and luciferase assay was used to validate the miRNA/target association. Western blot analysis and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) were used to determine the expression of miR‑23b‑3p and phosphatase and tensin homolog (PTEN), and their expression in smooth muscle cells (SMCs) treated with miRNA mimic or inhibitor. Firstly, an online miRNA database (www.mirdb.org) was searched using the 'seed sequence' located within the 3'‑untranslated region of the target gene, and then PTEN was validated as the direct target gene via a luciferase reporter assay system. The negative regulatory association between miR‑23b‑3p and PTEN was determined through the analysis of the relative luciferase activity. Additionally, RT-qPCR and western blot analysis was performed in order to assess the mRNA and protein expression levels of PTEN among IA (n=32) and control (n=17) groups or cells treated with scramble control, miR‑23b‑3p mimics, PTEN siRNA and miR‑23b‑3p inhibitors to verify the negative regulatory association between miR‑23b‑3p and PTEN. Experiments were then performed to investigate the effect of miR‑23b‑3p and PTEN on the viability and apoptosis of pulmonary artery SMCs (PASMCs). The results showed that cells transfected with miR‑23b‑3p inhibitors suppressed the viability of SMCs by promoting the apoptosis of the cells compared with that of the scramble controls, while cells transfected with miR‑23b‑3p mimics and PTEN siRNA enhanced the viability of VSMCs by inducing apoptosis. This indicated that miR‑23b‑3p negatively interfered with the viability of the cells, while PTEN positively interfered with the viability of the cells. In conclusion, PTEN was found to be a virtual target of miR‑23b‑3p, and a negative regulatory association existed between miR‑23b‑3p and PTEN. miR‑23b‑3p and PTEN interfered with the viability and apoptosis of SMCs.
Collapse
Affiliation(s)
- Dong Guo
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 410052, P.R. China
| | - Ye-Wei Wang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 410052, P.R. China
| | - Lei Yan
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 410052, P.R. China
| | - Ji Ma
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 410052, P.R. China
| | - Xin-Wei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 410052, P.R. China
| | - Shao-Feng Shui
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 410052, P.R. China
| |
Collapse
|
29
|
Zhang J, Yin J, Wang Y, Li B, Zeng X. Apelin impairs myogenic response to induce diabetic nephropathy in mice. FASEB J 2018. [PMID: 29522374 DOI: 10.1096/fj.201701257r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The cause of the invalid reaction of smooth muscle cells to mechanical stimulation that results in a dysfunctional myogenic response that mediates the disruption of renal blood flow (RBF) in patients with diabetes is debatable. The present study revealed that increased apelin concentration in serum of diabetic mice neutralized the myogenic response mediated by apelin receptor (APJ) and resulted in increased RBF, which promoted the progression of diabetic nephropathy. The results showed that apelin concentration, RBF, and albuminuria:creatinine ratio were all increased in kkAy mice, and increased RBF correlated positively with serum apelin both in C57 and diabetic mice. The increased RBF was accompanied by decreased phosphorylation of myosin light chain (MLC), β-arrestin, and increased endothelial NOS in glomeruli. Meanwhile, calcium, phosphorylation of MLC, and β-arrestin were decreased by high glucose and apelin treatment in cultured smooth muscle cells, as well. eNOS was increased by high glucose and increased by apelin in cultured endothelial cells (ECs). Knockdown of β-arrestin expression in smooth muscle cells cancelled phosphorylation of MLC induced by apelin. Therefore, apelin may induce the progression of diabetic nephropathy by counteracting the myogenic response in smooth muscle cells.-Zhang, J., Yin, J., Wang, Y., Li, B., Zeng, X. Apelin impairs myogenic response to induce diabetic nephropathy in mice.
Collapse
Affiliation(s)
- Jia Zhang
- Department of Pathophysiology, Capital Medical University, Beijing, China
| | - Jiming Yin
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing, China
| | - Yangjia Wang
- Department of Pathophysiology, Capital Medical University, Beijing, China
| | - Bin Li
- Department of Pathophysiology, Capital Medical University, Beijing, China
| | - Xiangjun Zeng
- Department of Pathophysiology, Capital Medical University, Beijing, China
| |
Collapse
|
30
|
Moulton KS, Li M, Strand K, Burgett S, McClatchey P, Tucker R, Furgeson SB, Lu S, Kirkpatrick B, Cleveland JC, Nemenoff RA, Ambardekar AV, Weiser-Evans MC. PTEN deficiency promotes pathological vascular remodeling of human coronary arteries. JCI Insight 2018; 3:97228. [PMID: 29467331 PMCID: PMC5916252 DOI: 10.1172/jci.insight.97228] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 01/23/2018] [Indexed: 01/20/2023] Open
Abstract
Phosphatase and tensin homolog (PTEN) is an essential regulator of the differentiated vascular smooth muscle cell (SMC) phenotype. Our goal was to establish that PTEN loss promotes SMC dedifferentiation and pathological vascular remodeling in human atherosclerotic coronary arteries and nonatherosclerotic coronary arteries exposed to continuous-flow left ventricular assist devices (CF-LVADs). Arteries were categorized as nonatherosclerotic hyperplasia (NAH), atherosclerotic hyperplasia (AH), or complex plaque (CP). NAH coronary arteries from CF-LVAD patients were compared to NAH coronaries from non-LVAD patients. Intimal PTEN and SMC contractile protein expression was reduced compared with the media in arteries with NAH, AH, or CP. Compared with NAH, PTEN and SMC contractile protein expression was reduced in the media and intima of arteries with AH and CP. NAH arteries from CF-LVAD patients showed marked vascular remodeling and reduced PTEN and α-smooth muscle actin (αSMA) in medial SMCs compared with arteries from non-LVAD patients; this correlated with increased medial collagen deposition. Mechanistically, compared with ApoE–/– mice, SMC-specific PTEN-null/ApoE–/– double-knockout mice exhibited accelerated atherosclerosis progression and increased vascular fibrosis. By microarray and validated quantitative RT-PCR analysis, SMC PTEN deficiency promotes a global upregulation of proinflammatory and profibrotic genes. We propose that PTEN is an antiinflammatory, antifibrotic target that functions to maintain SMC differentiation. SMC loss of PTEN results in pathological vascular remodeling of human arteries. PTEN loss correlates with dedifferentiation of smooth muscle cells of human coronary arteries affected with atherosclerosis or exposed to continuous-flow left ventricular assist devices.
Collapse
Affiliation(s)
| | - Marcella Li
- Division of Cardiology, Department of Medicine
| | - Keith Strand
- Division of Renal Diseases and Hypertension, Department of Medicine
| | - Shawna Burgett
- Division of Renal Diseases and Hypertension, Department of Medicine
| | | | - Rebecca Tucker
- Division of Renal Diseases and Hypertension, Department of Medicine
| | - Seth B Furgeson
- Division of Renal Diseases and Hypertension, Department of Medicine.,School of Medicine, Consortium for Fibrosis Research and Translation
| | - Sizhao Lu
- Division of Renal Diseases and Hypertension, Department of Medicine
| | | | - Joseph C Cleveland
- School of Medicine, Consortium for Fibrosis Research and Translation.,Department of Surgery
| | - Raphael A Nemenoff
- Division of Renal Diseases and Hypertension, Department of Medicine.,School of Medicine, Consortium for Fibrosis Research and Translation.,Cardiovascular Pulmonary Research Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Amrut V Ambardekar
- Division of Cardiology, Department of Medicine.,School of Medicine, Consortium for Fibrosis Research and Translation
| | - Mary Cm Weiser-Evans
- Division of Renal Diseases and Hypertension, Department of Medicine.,School of Medicine, Consortium for Fibrosis Research and Translation.,Cardiovascular Pulmonary Research Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
31
|
Roostalu U, Wong JK. Arterial smooth muscle dynamics in development and repair. Dev Biol 2018; 435:109-121. [PMID: 29397877 DOI: 10.1016/j.ydbio.2018.01.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Revised: 01/08/2018] [Accepted: 01/24/2018] [Indexed: 12/11/2022]
Abstract
Arterial vasculature distributes blood from early embryonic development and provides a nutrient highway to maintain tissue viability. Atherosclerosis, peripheral artery diseases, stroke and aortic aneurysm represent the most frequent causes of death and are all directly related to abnormalities in the function of arteries. Vascular intervention techniques have been established for the treatment of all of these pathologies, yet arterial surgery can itself lead to biological changes in which uncontrolled arterial wall cell proliferation leads to restricted blood flow. In this review we describe the intricate cellular composition of arteries, demonstrating how a variety of distinct cell types in the vascular walls regulate the function of arteries. We provide an overview of the developmental origin of arteries and perivascular cells and focus on cellular dynamics in arterial repair. We summarize the current knowledge of the molecular signaling pathways that regulate vascular smooth muscle differentiation in the embryo and in arterial injury response. Our review aims to highlight the similarities as well as differences between cellular and molecular mechanisms that control arterial development and repair.
Collapse
Affiliation(s)
- Urmas Roostalu
- Manchester Academic Health Science Centre, Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, UK.
| | - Jason Kf Wong
- Manchester Academic Health Science Centre, Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, UK; Department of Plastic Surgery, Manchester University NHS Foundation Trust, Wythenshawe Hospital, Manchester, UK.
| |
Collapse
|
32
|
Deng Y, Lin C, Zhou HJ, Min W. Smooth muscle cell differentiation: Mechanisms and models for vascular diseases. ACTA ACUST UNITED AC 2018. [DOI: 10.1007/s11515-017-1473-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
33
|
MicroRNA-20a participates in the aerobic exercise-based prevention of coronary artery disease by targeting PTEN. Biomed Pharmacother 2017; 95:756-763. [DOI: 10.1016/j.biopha.2017.08.086] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 08/11/2017] [Accepted: 08/23/2017] [Indexed: 12/25/2022] Open
|
34
|
Li X, Chen W, Li P, Wei J, Cheng Y, Liu P, Yan Q, Xu X, Cui Y, Gu Z, Simoncini T, Fu X. Follicular Stimulating Hormone Accelerates Atherogenesis by Increasing Endothelial VCAM-1 Expression. Theranostics 2017; 7:4671-4688. [PMID: 29187895 PMCID: PMC5706091 DOI: 10.7150/thno.21216] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Accepted: 09/08/2017] [Indexed: 01/02/2023] Open
Abstract
Rationale: Postmenopausal atherosclerosis (AS) has for decades been attributed to estrogen deficiency. Although the follicular stimulating hormone (FSH) levels rise sharply in parallel, the direct effect of FSH on AS has never been investigated. In this study, we explored the possible role of FSH in the development of AS. Methods: This was a prospective cohort study of 48 healthy premenopausal and 15 postmenopausal women. ApoE knockout mice were used as atherosclerosis model and human umbilical vascular endothelial cells (HUVECs) were cultured as cell model. Serum hormones and vascular cell adhesion molecule-1 (VCAM-1) levels were measured. Real-time PCR, histology for atherosclerotic lesions, immunofluorescence, luciferase assay, transfection experiments, flow chamber adhesion assay and western blot were performed. Results: In ApoE knockout mice, administration of FSH increased the atherosclerotic lesions and serum VCAM-1 concentration. Importantly, in blood samples of postmenopausal women, we detected significantly higher levels of FSH and VCAM-1 compared with those from premenopausal women, and there was a positive correlation between these two molecules. In cultured HUVECs, FSH receptor (FSHR) mRNA and protein expression were detected and FSH enhanced VCAM-1 expression. This effect was mediated by the activation of nuclear factor κB (NF-κB), which was sequentially enhanced by the activation of PI3K/Akt/mTOR cascade. FSH first enhanced GαS activity resulting in elevated cAMP level and PKA activity, which relayed the signals from FSHR to the PI3K/Akt/mTOR cascade. Furthermore, FSHR was detected in endothelial caveolae fraction and interacted with caveolin-1 and GαS. The disruption of caveolae or the silencing of caveolin-1 blocked FSH effects on signaling activation and VCAM-1 expression, suggesting the existence of a functional signaling module in membrane caveolae. Finally, FSH increased human monocyte adhesion to HUVECs which was reversed by the VCAM-1 neutralizing antibody. Conclusion: FSHR was located in the membrane caveolae of HUVECs and FSH promoted VCAM-1 expression via FSHR/GαS /cAMP/PKA and PI3K/Akt/mTOR/NF-κB pathway. This may contribute to the deleterious role of FSH in the development of AS in postmenopausal women.
Collapse
|
35
|
Liao L, Zheng B, Yi B, Liu C, Chen L, Zeng Z, Gao J. Annexin A2-modulated proliferation of pulmonary arterial smooth muscle cells depends on caveolae and caveolin-1 in hepatopulmonary syndrome. Exp Cell Res 2017; 359:266-274. [PMID: 28729092 DOI: 10.1016/j.yexcr.2017.07.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 07/14/2017] [Accepted: 07/15/2017] [Indexed: 11/25/2022]
Abstract
We have established that annexin A2 (ANXA2) is an important factor in the experimental hepatopulmonary syndrome (HPS) serum-induced proliferation of pulmonary arterial smooth muscle cells (PASMCs). However, the detailed mechanism remains unclear. ANXA2 translocated to the caveolin-enriched microdomains (caveolae) in PASMCs upon HPS serum stimulation. The disruption of caveolae by Methyl-β-cyclodextrin (MβCD) alleviated the caveolae recruitment of ANXA2 and the ANXA2-mediated activation of ERK1/2 and NF-κB, so that ANXA2-modulated PASMC proliferation was suppressed. The over-expression of Cav-1 resulted in the relocation of ANXA2 from caveolae and negatively regulated ERK1/2 and NF-κB activation, which inhibited the ANXA2-modulated PASMC proliferative behavior. These data indicate that caveolae function as a signaling platform for ANXA2-induced proliferative behavior and Cav-1 participates upstream of ANXA2 in the activation of ERK1/2 and NF-κB.
Collapse
Affiliation(s)
- Lin Liao
- Department of Anesthesia, People's Hospital of Qijiang District, Chongqing 401420, China
| | - Binwu Zheng
- Department of Anesthesia, People's Hospital of Rongchang County, Chongqing 402460, China
| | - Bin Yi
- Department of Anesthesia, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China
| | - Chang Liu
- Department of Anesthesia, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China
| | - Lin Chen
- Department of Anesthesia, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China
| | - Ziyang Zeng
- Department of Anesthesia, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China.
| | - Jing Gao
- Department of Anesthesia, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
36
|
Walker-Allgaier B, Schaub M, Alesutan I, Voelkl J, Geue S, Münzer P, Rodríguez JM, Kuhl D, Lang F, Gawaz M, Borst O. SGK1 up-regulates Orai1 expression and VSMC migration during neointima formation after arterial injury. Thromb Haemost 2017; 117:1002-1005. [PMID: 28203685 DOI: 10.1160/th16-09-0690] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 01/23/2017] [Indexed: 12/22/2022]
Abstract
Supplementary Material to this article is available online at www.thrombosis-online.com
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Meinrad Gawaz
- Meinrad Gawaz, MD, Department of Cardiology and Cardiovascular Medicine, University of Tübingen, Otfried Mueller-Str. 10, 72076 Tübingen, Germany, Tel.: +49 7071 2983688, Fax: +49 7071 294473 , E-mail:
| | - Oliver Borst
- Oliver Borst, MD, Department of Cardiology and Cardiovascular Medicine, University of Tübingen, Otfried Mueller-Str. 10, 72076 Tübingen, Germany, Tel.: +49 7071 2984483, Fax: +49 7071 294473, E-mail:
| |
Collapse
|
37
|
Long noncoding RNA H19-derived miR-675 aggravates restenosis by targeting PTEN. Biochem Biophys Res Commun 2017; 497:1154-1161. [PMID: 28063931 DOI: 10.1016/j.bbrc.2017.01.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 01/03/2017] [Indexed: 01/05/2023]
Abstract
Restenosis is mainly attributed to excessive proliferation of vascular smooth muscle cells (VSMCs). Noncoding RNAs have been identified as key regulators of diverse pathological processes. We reported that the long noncoding RNA H19 (LncRNA H19) and LncRNA H19-derived microRNA (miR-675) are overexpressed in neointima of balloon-injured artery. Thus, the present study aims to evaluate the role of LncRNA H19 on VSMCs proliferation. To determine the changes of LncRNA H19 and miR-675 expression in the injured arterial wall, the standard rat carotid artery balloon injury model was used. In vivo studies demonstrated that both LncRNA H19 and miR-675 were upregulated after vascular injury. Correlation analysis revealed a positive relationship between LncRNA H19/miR-675 and the ratio of intima to media. Gain-of-function studies showed that the overexpression of LncRNA H19 accelerated T/G HA-VSMC proliferation in vitro. We further validated that PTEN is the target gene of miR-675 as demonstrated by luciferase assay. Finally, the results of the rescue experiment indicated that LncRNA H19 promoted the proliferation of T/G HA-VSMC in a miR-675-dependent manner. This finding not only reveal a novel function of LncRNA H19, but also has important diagnostic and therapeutic implications in the setting of restenosis and perhaps other vascular proliferative disorders as well.
Collapse
|
38
|
Zhu Z, Zheng X, Li D, Wang T, Xu R, Piao H, Liu K. Prx1 promotes the proliferation and migration of vascular smooth muscle cells in a TLR4-dependent manner. Mol Med Rep 2016; 15:345-351. [DOI: 10.3892/mmr.2016.5987] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 08/02/2016] [Indexed: 11/06/2022] Open
|
39
|
Majesky MW, Horita H, Ostriker A, Lu S, Regan JN, Bagchi A, Dong XR, Poczobutt J, Nemenoff RA, Weiser-Evans MCM. Differentiated Smooth Muscle Cells Generate a Subpopulation of Resident Vascular Progenitor Cells in the Adventitia Regulated by Klf4. Circ Res 2016; 120:296-311. [PMID: 27834190 DOI: 10.1161/circresaha.116.309322] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 10/31/2016] [Accepted: 11/09/2016] [Indexed: 01/31/2023]
Abstract
RATIONALE The vascular adventitia is a complex layer of the vessel wall consisting of vasa vasorum microvessels, nerves, fibroblasts, immune cells, and resident progenitor cells. Adventitial progenitors express the stem cell markers, Sca1 and CD34 (adventitial sca1-positive progenitor cells [AdvSca1]), have the potential to differentiate in vitro into multiple lineages, and potentially contribute to intimal lesions in vivo. OBJECTIVE Although emerging data support the existence of AdvSca1 cells, the goal of this study was to determine their origin, degree of multipotency and heterogeneity, and contribution to vessel remodeling. METHODS AND RESULTS Using 2 in vivo fate-mapping approaches combined with a smooth muscle cell (SMC) epigenetic lineage mark, we report that a subpopulation of AdvSca1 cells is generated in situ from differentiated SMCs. Our data establish that the vascular adventitia contains phenotypically distinct subpopulations of progenitor cells expressing SMC, myeloid, and hematopoietic progenitor-like properties and that differentiated SMCs are a source to varying degrees of each subpopulation. SMC-derived AdvSca1 cells exhibit a multipotent phenotype capable of differentiating in vivo into mature SMCs, resident macrophages, and endothelial-like cells. After vascular injury, SMC-derived AdvSca1 cells expand in number and are major contributors to adventitial remodeling. Induction of the transcription factor Klf4 in differentiated SMCs is essential for SMC reprogramming in vivo, whereas in vitro approaches demonstrate that Klf4 is essential for the maintenance of the AdvSca1 progenitor phenotype. CONCLUSIONS We propose that generation of resident vascular progenitor cells from differentiated SMCs is a normal physiological process that contributes to the vascular stem cell pool and plays important roles in arterial homeostasis and disease.
Collapse
Affiliation(s)
- Mark W Majesky
- From the Division of Renal Diseases and Hypertension (H.H., A.O., S.L., A.B., J.P., R.A.N., M.C.M.W.-E.) and Cardiovascular Pulmonary Research Program, Division of Cardiology (R.A.N., M.C.M.W.-E.), School of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado, Aurora (M.C.M.W.-E.); Division of Endocrinology, Department of Medicine, Indiana University, Indianapolis (J.N.R.); Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, WA (M.W.M., X.R.D.); and Department of Pediatrics and Department of Pathology, University of Washington, Seattle (M.W.M.).
| | - Henrick Horita
- From the Division of Renal Diseases and Hypertension (H.H., A.O., S.L., A.B., J.P., R.A.N., M.C.M.W.-E.) and Cardiovascular Pulmonary Research Program, Division of Cardiology (R.A.N., M.C.M.W.-E.), School of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado, Aurora (M.C.M.W.-E.); Division of Endocrinology, Department of Medicine, Indiana University, Indianapolis (J.N.R.); Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, WA (M.W.M., X.R.D.); and Department of Pediatrics and Department of Pathology, University of Washington, Seattle (M.W.M.)
| | - Allison Ostriker
- From the Division of Renal Diseases and Hypertension (H.H., A.O., S.L., A.B., J.P., R.A.N., M.C.M.W.-E.) and Cardiovascular Pulmonary Research Program, Division of Cardiology (R.A.N., M.C.M.W.-E.), School of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado, Aurora (M.C.M.W.-E.); Division of Endocrinology, Department of Medicine, Indiana University, Indianapolis (J.N.R.); Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, WA (M.W.M., X.R.D.); and Department of Pediatrics and Department of Pathology, University of Washington, Seattle (M.W.M.)
| | - Sizhao Lu
- From the Division of Renal Diseases and Hypertension (H.H., A.O., S.L., A.B., J.P., R.A.N., M.C.M.W.-E.) and Cardiovascular Pulmonary Research Program, Division of Cardiology (R.A.N., M.C.M.W.-E.), School of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado, Aurora (M.C.M.W.-E.); Division of Endocrinology, Department of Medicine, Indiana University, Indianapolis (J.N.R.); Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, WA (M.W.M., X.R.D.); and Department of Pediatrics and Department of Pathology, University of Washington, Seattle (M.W.M.)
| | - Jenna N Regan
- From the Division of Renal Diseases and Hypertension (H.H., A.O., S.L., A.B., J.P., R.A.N., M.C.M.W.-E.) and Cardiovascular Pulmonary Research Program, Division of Cardiology (R.A.N., M.C.M.W.-E.), School of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado, Aurora (M.C.M.W.-E.); Division of Endocrinology, Department of Medicine, Indiana University, Indianapolis (J.N.R.); Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, WA (M.W.M., X.R.D.); and Department of Pediatrics and Department of Pathology, University of Washington, Seattle (M.W.M.)
| | - Ashim Bagchi
- From the Division of Renal Diseases and Hypertension (H.H., A.O., S.L., A.B., J.P., R.A.N., M.C.M.W.-E.) and Cardiovascular Pulmonary Research Program, Division of Cardiology (R.A.N., M.C.M.W.-E.), School of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado, Aurora (M.C.M.W.-E.); Division of Endocrinology, Department of Medicine, Indiana University, Indianapolis (J.N.R.); Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, WA (M.W.M., X.R.D.); and Department of Pediatrics and Department of Pathology, University of Washington, Seattle (M.W.M.)
| | - Xiu Rong Dong
- From the Division of Renal Diseases and Hypertension (H.H., A.O., S.L., A.B., J.P., R.A.N., M.C.M.W.-E.) and Cardiovascular Pulmonary Research Program, Division of Cardiology (R.A.N., M.C.M.W.-E.), School of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado, Aurora (M.C.M.W.-E.); Division of Endocrinology, Department of Medicine, Indiana University, Indianapolis (J.N.R.); Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, WA (M.W.M., X.R.D.); and Department of Pediatrics and Department of Pathology, University of Washington, Seattle (M.W.M.)
| | - Joanna Poczobutt
- From the Division of Renal Diseases and Hypertension (H.H., A.O., S.L., A.B., J.P., R.A.N., M.C.M.W.-E.) and Cardiovascular Pulmonary Research Program, Division of Cardiology (R.A.N., M.C.M.W.-E.), School of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado, Aurora (M.C.M.W.-E.); Division of Endocrinology, Department of Medicine, Indiana University, Indianapolis (J.N.R.); Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, WA (M.W.M., X.R.D.); and Department of Pediatrics and Department of Pathology, University of Washington, Seattle (M.W.M.)
| | - Raphael A Nemenoff
- From the Division of Renal Diseases and Hypertension (H.H., A.O., S.L., A.B., J.P., R.A.N., M.C.M.W.-E.) and Cardiovascular Pulmonary Research Program, Division of Cardiology (R.A.N., M.C.M.W.-E.), School of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado, Aurora (M.C.M.W.-E.); Division of Endocrinology, Department of Medicine, Indiana University, Indianapolis (J.N.R.); Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, WA (M.W.M., X.R.D.); and Department of Pediatrics and Department of Pathology, University of Washington, Seattle (M.W.M.)
| | - Mary C M Weiser-Evans
- From the Division of Renal Diseases and Hypertension (H.H., A.O., S.L., A.B., J.P., R.A.N., M.C.M.W.-E.) and Cardiovascular Pulmonary Research Program, Division of Cardiology (R.A.N., M.C.M.W.-E.), School of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado, Aurora (M.C.M.W.-E.); Division of Endocrinology, Department of Medicine, Indiana University, Indianapolis (J.N.R.); Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, WA (M.W.M., X.R.D.); and Department of Pediatrics and Department of Pathology, University of Washington, Seattle (M.W.M.).
| |
Collapse
|
40
|
Zhang MJ, Liu Y, Hu ZC, Zhou Y, Pi Y, Guo L, Wang X, Chen X, Li JC, Zhang LL. TRPV1 attenuates intracranial arteriole remodeling through inhibiting VSMC phenotypic modulation in hypertension. Histochem Cell Biol 2016; 147:511-521. [DOI: 10.1007/s00418-016-1512-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2016] [Indexed: 01/11/2023]
|
41
|
Daniel JM, Reichel CA, Schmidt-Woell T, Dutzmann J, Zuchtriegel G, Krombach F, Herold J, Bauersachs J, Sedding DG, Kanse SM. Factor VII-activating protease deficiency promotes neointima formation by enhancing leukocyte accumulation. J Thromb Haemost 2016; 14:2058-2067. [PMID: 27431088 DOI: 10.1111/jth.13417] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 07/01/2016] [Indexed: 01/08/2023]
Abstract
Essentials Factor VII-activating protease (FSAP) is a plasma protease involved in vascular processes. Neointima formation was investigated after vascular injury in FSAP-/- mice. The neointimal lesion size and the accumulation of macrophages were increased in FSAP-/- mice. This was due to an increased activity of the chemokine (C-C motif) ligand 2 (CCL2). SUMMARY Background Factor VII-activating protease (FSAP) is a multifunctional circulating plasma serine protease involved in thrombosis and vascular remodeling processes. The Marburg I single-nucleotide polymorphism (MI-SNP) in the FSAP-coding gene is characterized by low proteolytic activity, and is associated with increased rates of stroke and carotid stenosis in humans. Objectives To determine whether neointima formation after vascular injury is increased in FSAP-/- mice. Methods and Results The neointimal lesion size and the proliferation of vascular smooth muscle cells (VSMCs) were significantly enhanced in FSAP-/- mice as compared with C57BL/6 control mice after wire-induced injury of the femoral artery. Accumulation of leukocytes and macrophages was increased within the lesions of FSAP-/- mice at day 3 and day 14. Quantitative zymography demonstrated enhanced activity of gelatinases/matrix metalloproteinase (MMP)-2 and MMP-9 within the neointimal lesions of FSAP-/- mice, and immunohistochemistry showed particular costaining of MMP-9 with accumulating leukocytes. Using intravital microscopy, we observed that FSAP deficiency promoted the intravascular adherence and the subsequent transmigration of leukocytes in vivo in response to chemokine ligand 2 (CCL2). CCL2 expression was increased in FSAP-/- monocytes but not in the vessel wall. There was no difference in the expression of platelet-derived growth factor (PDGF-BB). Conclusions FSAP deficiency causes an increase in CCL2 expression and CCL2-mediated infiltration of leukocytes into the injured vessel, thereby promoting SMC proliferation and migration by the activation of leukocyte-derived gelatinases. These results provide a possible explanation for the observed association of the loss-of-function MI-SNP with vascular proliferative diseases.
Collapse
Affiliation(s)
- J-M Daniel
- Department of Biochemistry, Justus-Liebig-Universität Giessen, Giessen, Germany
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - C A Reichel
- Department of Otorhinolaryngology, Head and Neck Surgery, Ludwig-Maximilians-Universität München, Munich, Germany
- Walter Brendel Center of Experimental Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - T Schmidt-Woell
- Department of Biochemistry, Justus-Liebig-Universität Giessen, Giessen, Germany
| | - J Dutzmann
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - G Zuchtriegel
- Department of Otorhinolaryngology, Head and Neck Surgery, Ludwig-Maximilians-Universität München, Munich, Germany
- Walter Brendel Center of Experimental Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - F Krombach
- Walter Brendel Center of Experimental Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - J Herold
- Department of Cardiology, Angiology and Pneumology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - J Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - D G Sedding
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - S M Kanse
- Department of Biochemistry, Justus-Liebig-Universität Giessen, Giessen, Germany.
- Oslo University Hospital and University of Oslo, Oslo, Norway.
| |
Collapse
|
42
|
Frismantiene A, Dasen B, Pfaff D, Erne P, Resink TJ, Philippova M. T-cadherin promotes vascular smooth muscle cell dedifferentiation via a GSK3β-inactivation dependent mechanism. Cell Signal 2016; 28:516-530. [DOI: 10.1016/j.cellsig.2016.02.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 02/12/2016] [Accepted: 02/18/2016] [Indexed: 11/24/2022]
|
43
|
Nuclear PTEN functions as an essential regulator of SRF-dependent transcription to control smooth muscle differentiation. Nat Commun 2016; 7:10830. [PMID: 26940659 PMCID: PMC5411712 DOI: 10.1038/ncomms10830] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 01/25/2016] [Indexed: 12/13/2022] Open
Abstract
Vascular disease progression is associated with marked changes in vascular smooth muscle cell (SMC) phenotype and function. SMC contractile gene expression and, thus differentiation, is under direct transcriptional control by the transcription factor, serum response factor (SRF); however, the mechanisms dynamically regulating SMC phenotype are not fully defined. Here we report that the lipid and protein phosphatase, PTEN, has a novel role in the nucleus by functioning as an indispensible regulator with SRF to maintain the differentiated SM phenotype. PTEN interacts with the N-terminal domain of SRF and PTEN–SRF interaction promotes SRF binding to essential promoter elements in SM-specific genes. Factors inducing phenotypic switching promote loss of nuclear PTEN through nucleo-cytoplasmic translocation resulting in reduced myogenically active SRF, but enhanced SRF activity on target genes involved in proliferation. Overall decreased expression of PTEN was observed in intimal SMCs of human atherosclerotic lesions underlying the potential clinical importance of these findings. The transcription factor, serum response factor, SRF regulates critical smooth muscle (SM) contractile gene expression but what else controls SM differentiation is unclear. Here, Horita et al. demonstrate that nuclear PTEN acts with SRF at the transcriptional level to maintain the differentiated SM phenotype.
Collapse
|
44
|
Gavin KM, Gutman JA, Kohrt WM, Wei Q, Shea KL, Miller HL, Sullivan TM, Erickson PF, Helm KM, Acosta AS, Childs CR, Musselwhite E, Varella-Garcia M, Kelly K, Majka SM, Klemm DJ. De novo generation of adipocytes from circulating progenitor cells in mouse and human adipose tissue. FASEB J 2015; 30:1096-108. [PMID: 26581599 DOI: 10.1096/fj.15-278994] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 11/02/2015] [Indexed: 12/21/2022]
Abstract
White adipocytes in adults are typically derived from tissue resident mesenchymal progenitors. The recent identification of de novo production of adipocytes from bone marrow progenitor-derived cells in mice challenges this paradigm and indicates an alternative lineage specification that adipocytes exist. We hypothesized that alternative lineage specification of white adipocytes is also present in human adipose tissue. Bone marrow from transgenic mice in which luciferase expression is governed by the adipocyte-restricted adiponectin gene promoter was adoptively transferred to wild-type recipient mice. Light emission was quantitated in recipients by in vivo imaging and direct enzyme assay. Adipocytes were also obtained from human recipients of hematopoietic stem cell transplantation. DNA was isolated, and microsatellite polymorphisms were exploited to quantify donor/recipient chimerism. Luciferase emission was detected from major fat depots of transplanted mice. No light emission was observed from intestines, liver, or lungs. Up to 35% of adipocytes in humans were generated from donor marrow cells in the absence of cell fusion. Nontransplanted mice and stromal-vascular fraction samples were used as negative and positive controls for the mouse and human experiments, respectively. This study provides evidence for a nontissue resident origin of an adipocyte subpopulation in both mice and humans.
Collapse
Affiliation(s)
- Kathleen M Gavin
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Jonathan A Gutman
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Wendy M Kohrt
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Qi Wei
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Karen L Shea
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Heidi L Miller
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Timothy M Sullivan
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Paul F Erickson
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Karen M Helm
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Alistaire S Acosta
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Christine R Childs
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Evelyn Musselwhite
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Marileila Varella-Garcia
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Kimberly Kelly
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Susan M Majka
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Dwight J Klemm
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
45
|
Saddouk FZ, Sun LY, Liu YF, Jiang M, Singer DV, Backs J, Van Riper D, Ginnan R, Schwarz JJ, Singer HA. Ca2+/calmodulin-dependent protein kinase II-γ (CaMKIIγ) negatively regulates vascular smooth muscle cell proliferation and vascular remodeling. FASEB J 2015; 30:1051-64. [PMID: 26567004 DOI: 10.1096/fj.15-279158] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 10/28/2015] [Indexed: 01/15/2023]
Abstract
Vascular smooth muscle (VSM) expresses calcium/calmodulin-dependent protein kinase II (CaMKII)-δ and -γ isoforms. CaMKIIδ promotes VSM proliferation and vascular remodeling. We tested CaMKIIγ function in vascular remodeling after injury. CaMKIIγ protein decreased 90% 14 d after balloon injury in rat carotid artery. Intraluminal transduction of adenovirus encoding CaMKIIγC rescued expression to 35% of uninjured controls, inhibited neointima formation (>70%), inhibited VSM proliferation (>60%), and increased expression of the cell-cycle inhibitor p21 (>2-fold). Comparable doses of CaMKIIδ2 adenovirus had no effect. Similar dynamics in CaMKIIγ mRNA and protein expression were observed in ligated mouse carotid arteries, correlating closely with expression of VSM differentiation markers. Targeted deletion of CaMKIIγ in smooth muscle resulted in a 20-fold increase in neointimal area, with a 3-fold increase in the cell proliferation index, no change in apoptosis, and a 60% decrease in p21 expression. In cultured VSM, CaMKIIγ overexpression induced p53 mRNA (1.7 fold) and protein (1.8-fold) expression; induced the p53 target gene p21 (3-fold); decreased VSM cell proliferation (>50%); and had no effect on expression of apoptosis markers. We conclude that regulated CaMKII isoform composition is an important determinant of the injury-induced vasculoproliferative response and that CaMKIIγ and -δ isoforms have nonequivalent, opposing functions.
Collapse
Affiliation(s)
- Fatima Z Saddouk
- *Center for Cardiovascular Sciences, Albany Medical College, Albany, New York, USA; and Department of Cardiology, Angiology and Pneumology, University of Heidelberg, Heidelberg, Germany
| | - Li-Yan Sun
- *Center for Cardiovascular Sciences, Albany Medical College, Albany, New York, USA; and Department of Cardiology, Angiology and Pneumology, University of Heidelberg, Heidelberg, Germany
| | - Yong Feng Liu
- *Center for Cardiovascular Sciences, Albany Medical College, Albany, New York, USA; and Department of Cardiology, Angiology and Pneumology, University of Heidelberg, Heidelberg, Germany
| | - Miao Jiang
- *Center for Cardiovascular Sciences, Albany Medical College, Albany, New York, USA; and Department of Cardiology, Angiology and Pneumology, University of Heidelberg, Heidelberg, Germany
| | - Diane V Singer
- *Center for Cardiovascular Sciences, Albany Medical College, Albany, New York, USA; and Department of Cardiology, Angiology and Pneumology, University of Heidelberg, Heidelberg, Germany
| | - Johannes Backs
- *Center for Cardiovascular Sciences, Albany Medical College, Albany, New York, USA; and Department of Cardiology, Angiology and Pneumology, University of Heidelberg, Heidelberg, Germany
| | - Dee Van Riper
- *Center for Cardiovascular Sciences, Albany Medical College, Albany, New York, USA; and Department of Cardiology, Angiology and Pneumology, University of Heidelberg, Heidelberg, Germany
| | - Roman Ginnan
- *Center for Cardiovascular Sciences, Albany Medical College, Albany, New York, USA; and Department of Cardiology, Angiology and Pneumology, University of Heidelberg, Heidelberg, Germany
| | - John J Schwarz
- *Center for Cardiovascular Sciences, Albany Medical College, Albany, New York, USA; and Department of Cardiology, Angiology and Pneumology, University of Heidelberg, Heidelberg, Germany
| | - Harold A Singer
- *Center for Cardiovascular Sciences, Albany Medical College, Albany, New York, USA; and Department of Cardiology, Angiology and Pneumology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
46
|
Yu D, Makkar G, Strickland DK, Blanpied TA, Stumpo DJ, Blackshear PJ, Sarkar R, Monahan TS. Myristoylated Alanine-Rich Protein Kinase Substrate (MARCKS) Regulates Small GTPase Rac1 and Cdc42 Activity and Is a Critical Mediator of Vascular Smooth Muscle Cell Migration in Intimal Hyperplasia Formation. J Am Heart Assoc 2015; 4:e002255. [PMID: 26450120 PMCID: PMC4845127 DOI: 10.1161/jaha.115.002255] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Transcription of the myristoylated alanine-rich C kinase substrate (MARCKS) is upregulated in animal models of intimal hyperplasia. MARCKS knockdown inhibits vascular smooth muscle cell (VSMC) migration in vitro; however, the mechanism is as yet unknown. We sought to elucidate the mechanism of MARCKS-mediated motility and determine whether MARCKS knockdown reduces intimal hyperplasia formation in vivo. METHODS AND RESULTS MARCKS knockdown blocked platelet-derived growth factor (PDGF)-induced translocation of cortactin to the cell cortex, impaired both lamellipodia and filopodia formation, and attenuated motility of human coronary artery smooth muscle cells (CASMCs). Activation of the small GTPases, Rac1 and Cdc42, was prevented by MARCKS knockdown. Phosphorylation of MARCKS resulted in a transient shift of MARCKS from the plasma membrane to the cytosol. MARCKS knockdown significantly decreased membrane-associated phosphatidylinositol 4,5-bisphosphate (PIP2) levels. Cotransfection with an intact, unphosphorylated MARCKS, which has a high binding affinity for PIP2, restored membrane-associated PIP2 levels and was indispensable for activation of Rac1 and Cdc42 and, ultimately, VSMC migration. Overexpression of MARCKS in differentiated VSMCs increased membrane PIP2 abundance, Rac1 and Cdc42 activity, and cell motility. MARCKS protein was upregulated early in the development of intimal hyperplasia in the murine carotid ligation model. Decreased MARKCS expression, but not total knockdown, attenuated intimal hyperplasia formation. CONCLUSIONS MARCKS upregulation increases VSMC motility by activation of Rac1 and Cdc42. These effects are mediated by MARCKS sequestering PIP2 at the plasma membrane. This study delineates a novel mechanism for MARCKS-mediated VSMC migration and supports the rational for MARCKS knockdown to prevent intimal hyperplasia.
Collapse
Affiliation(s)
- Dan Yu
- Department of Surgery, Veterans Affairs Medical Center, Baltimore, MD (D.Y., T.S.M.) Department of Surgery, University of Maryland School of Medicine, Baltimore, MD (D.Y., G.M., D.K.S., R.S., T.S.M.) Center for Vascular and Inflammatory Disease, University of Maryland School of Medicine, Baltimore, MD (D.Y., D.K.S., R.S., T.S.M.)
| | - George Makkar
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD (D.Y., G.M., D.K.S., R.S., T.S.M.)
| | - Dudley K Strickland
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD (D.Y., G.M., D.K.S., R.S., T.S.M.) Department of Physiology, University of Maryland School of Medicine, Baltimore, MD (D.K.S., T.A.B., R.S.) Center for Vascular and Inflammatory Disease, University of Maryland School of Medicine, Baltimore, MD (D.Y., D.K.S., R.S., T.S.M.)
| | - Thomas A Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD (D.K.S., T.A.B., R.S.)
| | - Deborah J Stumpo
- The Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC (D.J.S., P.J.B.)
| | - Perry J Blackshear
- The Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC (D.J.S., P.J.B.)
| | - Rajabrata Sarkar
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD (D.Y., G.M., D.K.S., R.S., T.S.M.) Department of Physiology, University of Maryland School of Medicine, Baltimore, MD (D.K.S., T.A.B., R.S.) Center for Vascular and Inflammatory Disease, University of Maryland School of Medicine, Baltimore, MD (D.Y., D.K.S., R.S., T.S.M.)
| | - Thomas S Monahan
- Department of Surgery, Veterans Affairs Medical Center, Baltimore, MD (D.Y., T.S.M.) Department of Surgery, University of Maryland School of Medicine, Baltimore, MD (D.Y., G.M., D.K.S., R.S., T.S.M.) Center for Vascular and Inflammatory Disease, University of Maryland School of Medicine, Baltimore, MD (D.Y., D.K.S., R.S., T.S.M.)
| |
Collapse
|
47
|
Zhang Y, Liu D, Wang L, Wang S, Yu X, Dai E, Liu X, Luo S, Jiang W. Integrated systems approach identifies risk regulatory pathways and key regulators in coronary artery disease. J Mol Med (Berl) 2015. [PMID: 26208504 DOI: 10.1007/s00109-015-1315-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
UNLABELLED Coronary artery disease (CAD) is the most common type of heart disease. However, the molecular mechanisms of CAD remain elusive. Regulatory pathways are known to play crucial roles in many pathogenic processes. Thus, inferring risk regulatory pathways is an important step toward elucidating the mechanisms underlying CAD. With advances in high-throughput data, we developed an integrated systems approach to identify CAD risk regulatory pathways and key regulators. Firstly, a CAD-related core subnetwork was identified from a curated transcription factor (TF) and microRNA (miRNA) regulatory network based on a random walk algorithm. Secondly, candidate risk regulatory pathways were extracted from the subnetwork by applying a breadth-first search (BFS) algorithm. Then, risk regulatory pathways were prioritized based on multiple CAD-associated data sources. Finally, we also proposed a new measure to prioritize upstream regulators. We inferred that phosphatase and tensin homolog (PTEN) may be a key regulator in the dysregulation of risk regulatory pathways. This study takes a closer step than the identification of disease subnetworks or modules. From the risk regulatory pathways, we could understand the flow of regulatory information in the initiation and progression of the disease. Our approach helps to uncover its potential etiology. KEY MESSAGES We developed an integrated systems approach to identify risk regulatory pathways. We proposed a new measure to prioritize the key regulators in CAD. PTEN may be a key regulator in dysregulation of the risk regulatory pathways.
Collapse
Affiliation(s)
- Yan Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Dianming Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Lihong Wang
- Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin, China
| | - Shuyuan Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xuexin Yu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Enyu Dai
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xinyi Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Shanshun Luo
- Department of Gerontology, The First Hospital of Harbin Medical University, Harbin Medical University, Harbin, China.
| | - Wei Jiang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China.
| |
Collapse
|
48
|
Huang XZ, Liu XX, Song JT, Wang L, Liu XL, Qu HY, Wang SX, Zhang C, Zhang Y, Zhang M. Prolyl-4-hydroxylase- 1 improves the stability of advanced plaques but accelerates the atherosclerotic lesion formation of early plaques. Eur Heart J Suppl 2015. [DOI: 10.1093/eurheartj/suv028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
49
|
Chistiakov DA, Orekhov AN, Bobryshev YV. Vascular smooth muscle cell in atherosclerosis. Acta Physiol (Oxf) 2015; 214:33-50. [PMID: 25677529 DOI: 10.1111/apha.12466] [Citation(s) in RCA: 292] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 02/02/2015] [Accepted: 02/09/2015] [Indexed: 12/30/2022]
Abstract
Vascular smooth muscle cells (VSMCs) exhibit phenotypic and functional plasticity in order to respond to vascular injury. In case of the vessel damage, VSMCs are able to switch from the quiescent 'contractile' phenotype to the 'proinflammatory' phenotype. This change is accompanied by decrease in expression of smooth muscle (SM)-specific markers responsible for SM contraction and production of proinflammatory mediators that modulate induction of proliferation and chemotaxis. Indeed, activated VSMCs could efficiently proliferate and migrate contributing to the vascular wall repair. However, in chronic inflammation that occurs in atherosclerosis, arterial VSMCs become aberrantly regulated and this leads to increased VSMC dedifferentiation and extracellular matrix formation in plaque areas. Proatherosclerotic switch in VSMC phenotype is a complex and multistep mechanism that may be induced by a variety of proinflammatory stimuli and hemodynamic alterations. Disturbances in hemodynamic forces could initiate the proinflammatory switch in VSMC phenotype even in pre-clinical stages of atherosclerosis. Proinflammatory signals play a crucial role in further dedifferentiation of VSMCs in affected vessels and propagation of pathological vascular remodelling.
Collapse
Affiliation(s)
- D. A. Chistiakov
- Research Center for Children's Health; Moscow Russia
- The Mount Sinai Community Clinical Oncology Program; Mount Sinai Comprehensive Cancer Center; Mount Sinai Medical Center; Miami Beach FL USA
| | - A. N. Orekhov
- Institute for Atherosclerosis; Skolkovo Innovative Center; Moscow Russia
- Laboratory of Angiopathology; Institute of General Pathology and Pathophysiology; Russian Academy of Sciences; Moscow Russia
- Department of Biophysics; Biological Faculty; Moscow State University; Moscow Russia
| | - Y. V. Bobryshev
- Institute for Atherosclerosis; Skolkovo Innovative Center; Moscow Russia
- Faculty of Medicine; School of Medical Sciences; University of New South Wales; Kensington Sydney NSW Australia
- School of Medicine; University of Western Sydney; Campbelltown NSW Australia
| |
Collapse
|
50
|
Yu XH, Zheng XL, Tang CK. Nuclear Factor-κB Activation as a Pathological Mechanism of Lipid Metabolism and Atherosclerosis. Adv Clin Chem 2015; 70:1-30. [PMID: 26231484 DOI: 10.1016/bs.acc.2015.03.004] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial wall with lipid-laden lesions, involving a complex interaction between multiple different cell types and cytokine networks. Inflammatory responses mark all stages of atherogenesis: from lipid accumulation in the intima to plaque formation and eventual rupture. One of the most important regulators of inflammation is the transcription factor nuclear factor-κB (NF-κB), which is activated through the canonical and noncanonical pathways in response to various stimuli. NF-κB has long been regarded as a proatherogenic factor, because it is implicated in multiple pathological processes during atherogenesis, including foam cell formation, vascular inflammation, proliferation of vascular smooth muscle cells, arterial calcification, and plaque progression. In contrast, inhibition of NF-κB signaling has been shown to protect against atherosclerosis. This chapter aims to discuss recent progress on the roles of NF-κB in lipid metabolism and atherosclerosis and also to highlight its potential therapeutic benefits.
Collapse
Affiliation(s)
- Xiao-Hua Yu
- Key Laboratory for Atherosclerology of Hunan Province, Molecular Target New Drug Discovery and Cooperative Innovation Center of Hunan Province, Life Science Research Center, University of South China, Hengyang, PR China
| | - Xi-Long Zheng
- Department of Biochemistry and Molecular Biology, The Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, The University of Calgary, Health Sciences Center, Calgary, Alberta, Canada
| | - Chao-Ke Tang
- Key Laboratory for Atherosclerology of Hunan Province, Molecular Target New Drug Discovery and Cooperative Innovation Center of Hunan Province, Life Science Research Center, University of South China, Hengyang, PR China.
| |
Collapse
|